51
|
Massey RS, Johri S, Chan D, Holahan MR, Prakash R. Comparison of Aptamer and Antibody Bioreceptors in the OEGFET Biosensor Platform for Detecting α-Synuclein, a Parkinson's Biomarker. IEEE SENSORS LETTERS 2024; 8:1-4. [DOI: 10.1109/lsens.2024.3413575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Roslyn S. Massey
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Srishti Johri
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| | - Dennis Chan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Matthew R. Holahan
- Department of Neuroscience, Health Sciences Building, Carleton University, Ottawa, ON, Canada
| | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
52
|
Wang Z, Gilliland T, Kim HJ, Gerasimenko M, Sajewski K, Camacho MV, Bebek G, Chen SG, Gunzler SA, Kong Q. A minimally Invasive Biomarker for Sensitive and Accurate Diagnosis of Parkinson's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.29.24309703. [PMID: 38978648 PMCID: PMC11230335 DOI: 10.1101/2024.06.29.24309703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Importance Parkinson's disease (PD), the second most common neurodegenerative disease, is pathologically characterized by intraneuronal deposition of misfolded alpha-synuclein aggregates (αSyn D ). αSyn D seeding activities in CSF and skin samples have shown great promise in PD diagnosis, but they require invasive procedures. Sensitive and accurate αSyn D seed amplification assay (αSyn-SAA) for more accessible and minimally invasive samples (such as blood and saliva) are urgently needed for PD pathological diagnosis in routine clinical practice. Objective To develop a sensitive and accurate αSyn-SAA biomarker using blood and saliva samples for sensitive, accurate and minimally invasive PD diagnosis. Design Setting and Participants This prospective diagnostic study evaluates serum and saliva samples collected from patients clinically diagnosed with PD or healthy controls (HC) without PD at an academic Parkinson's and Movement Disorders Center from February 2020 to March 2024. Patients diagnosed with non-PD parkinsonism were excluded from this analysis. A total of 124 serum samples (82 PD and 42 HC) and 131 saliva samples (83 PD and 48 HC) were collected and examined by αSyn-SAA. Out of the 124 serum donors, a subset of 74 subjects (48 PD and 26 HC) also donated saliva samples during the same visits. PD patients with serum samples had a mean age of 69.21 years (range 44-88); HC subjects with serum samples had a mean age of 66.55 years (range 44-81); PD patients with saliva samples had a mean age of 69.58 years (range 49-87); HC subjects with saliva samples had a mean age of 64.71 years (range 30-81). Main Outcomes and Measures Serum and/or saliva αSyn D seeding activities from PD and HC subjects were measured by αSyn-SAA using the Real-Time Quaking-Induced Conversion (RT-QuIC) platform. These PD patients had extensive clinical assessments including MDS-UPDRS. For a subset of PD and HC subjects whose serum and saliva samples were both collected during the same visits, the αSyn D seeding activities in both samples from the same subjects were examined, and the diagnostic accuracies for PD based on the seeding activities in either sample alone or both samples together were compared. Results RT-QuIC analysis of αSyn D seeding activities in the 124 serum samples revealed a sensitivity of 80.49%, a specificity of 90.48%, and an accuracy of 0.9006 (AUC of ROC, 95% CI, 0.8472-0.9539, p <0.0001) for PD diagnosis. RT-QuIC analysis of αSyn D seeding activity in 131 saliva samples revealed a sensitivity of 74.70%, a specificity of 97.92%, and an accuracy of 0.8966 (AUC of ROC, 95% CI, 0.8454-0.9478, p <0.0001). When aSyn D seeding activities in the paired serum-saliva samples from the subset of 48 PD and 26 HC subjects were considered together, sensitivity was 95.83%, specificity was 96.15%, and the accuracy was 0.98 (AUC of ROC, 95% CI, 0.96-1.00, p <0.001), which are significantly better than when αSyn D seeding activities in either serum or saliva were used alone. For the paired serum-saliva samples, when specificity was set at 100% by elevating the αSyn-SAA cutoff values, a sensitivity of 91.7% and an accuracy of 0.9457 were still attained. Detailed correlation analysis revealed that αSyn D seeding activities in the serum of PD patients were correlated inversely with Montreal Cognitive Assessment (MoCA) score ( p =0.04), positively with Hamilton Depression Rating Scale (HAM-D) ( p =0.03), and weakly positively with PDQ-39 cognitive impairment score ( p =0.07). Subgroup analysis revealed that the inverse correlation with MoCA was only seen in males ( p =0.013) and weakly in the ≥70 age group ( p =0.07), and that the positive correlation with HAM-D was only seen in females ( p =0.04) and in the <70 age group ( p =0.01). In contrast, αSyn D seeding activities in the saliva of PD patients were inversely correlated with age at diagnosis ( p =0.02) and the REM sleep behavior disorder (RBD) status ( p =0.04), but subgroup analysis showed that the inverse correlation with age at diagnosis was only seen in males ( p =0.04) and in the <70 age group ( p =0.01). Conclusion and Relevance Our data show that concurrent RT-QuIC assay of αSyn D seeding activities in both serum and saliva can achieve high diagnostic accuracies comparable to that of CSF αSyn-SAA, suggesting that αSyn D seeding activities in serum and saliva together can potentially be used as a valuable biomarker for highly sensitive, accurate, and minimally invasive diagnosis of PD in routine clinical practice. αSyn D seeding activities in serum and saliva of PD patients correlate differentially with some clinical characteristics and in an age and sex-dependent manner. KEY POINTS Question: Are αSyn D seeding activities in serum and saliva together a more sensitive and accurate diagnostic PD biomarker than αSyn D seeding activities in either sample type alone? Are αSyn D seeding activities in either serum or saliva correlated with any clinical characteristics? Findings: Examinations of αSyn D seeding activities in 124 serum samples and 131 saliva samples from PD and heathy control subjects show that αSyn D seeding activities in both serum and saliva samples together can provide significantly more sensitive and accurate diagnosis of PD than either sample type alone. αSyn D seeding activities in serum or saliva exhibit varied inverse or positive correlations with some clinical features in an age and sex-dependent manner. Meaning: αSyn D seeding activities in serum and saliva together can potentially be used as a valuable pathological biomarker for highly sensitive, accurate, and minimally invasive PD diagnosis in routine clinical practice and clinical studies, and αSyn D seeding activities in serum or saliva correlate with some clinical characteristics in an age and sex-dependent manner, suggesting some possible clinical utility of quantitative serum/saliva αSyn-SAA data.
Collapse
|
53
|
Di Luca DG, Perlmutter JS. Time for Clinical Dopamine Transporter Scans in Parkinsonism?: Not DAT Yet. Neurology 2024; 102:e209558. [PMID: 38759140 PMCID: PMC11175627 DOI: 10.1212/wnl.0000000000209558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 05/19/2024] Open
Affiliation(s)
- Daniel G Di Luca
- From the Department of Neurology, Washington University in St. Louis, MO
| | - Joel S Perlmutter
- From the Department of Neurology, Washington University in St. Louis, MO
| |
Collapse
|
54
|
Janarthanam C, Clabaugh G, Wang Z, Melvin BR, Scheibe I, Jin H, Anantharam V, Urbauer RJB, Urbauer JL, Ma J, Kanthasamy A, Huang X, Donadio V, Zou W, Kanthasamy AG. High-Yield α-Synuclein Purification and Ionic Strength Modification Pivotal to Seed Amplification Assay Performance and Reproducibility. Int J Mol Sci 2024; 25:5988. [PMID: 38892177 PMCID: PMC11172462 DOI: 10.3390/ijms25115988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Alpha-synuclein seed amplification assays (αSyn-SAAs) have emerged as promising diagnostic tools for Parkinson's disease (PD) by detecting misfolded αSyn and amplifying the signal through cyclic shaking and resting in vitro. Recently, our group and others have shown that multiple biospecimens, including CSF, skin, and submandibular glands (SMGs), can be used to seed the aggregation reaction and robustly distinguish between patients with PD and non-disease controls. The ultrasensitivity of the assay affords the ability to detect minute quantities of αSyn in peripheral tissues, but it also produces various technical challenges of variability. To address the problem of variability, we present a high-yield αSyn protein purification protocol for the efficient production of monomers with a low propensity for self-aggregation. We expressed wild-type αSyn in BL21 Escherichia coli, lysed the cells using osmotic shock, and isolated αSyn using acid precipitation and fast protein liquid chromatography (FPLC). Following purification, we optimized the ionic strength of the reaction buffer to distinguish the fluorescence maximum (Fmax) separation between disease and healthy control tissues for enhanced assay performance. Our protein purification protocol yielded high quantities of αSyn (average: 68.7 mg/mL per 1 L of culture) and showed highly precise and robust αSyn-SAA results using brain, skin, and SMGs with inter-lab validation.
Collapse
Affiliation(s)
- Chelva Janarthanam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Griffin Clabaugh
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Zerui Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Bradley R. Melvin
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA;
| | - Ileia Scheibe
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Huajun Jin
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Vellareddy Anantharam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Ramona J. B. Urbauer
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (R.J.B.U.); (J.L.U.)
| | - Jeffrey L. Urbauer
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (R.J.B.U.); (J.L.U.)
| | - Jiyan Ma
- Chinese Institute for Brain Research, Beijing 102206, China;
| | - Arthi Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences of Bologna, Complex Operational Unit Clinica Neurologica, 40138 Bologna, Italy;
| | - Wenquan Zou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Anumantha G. Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| |
Collapse
|
55
|
Mulroy E, Erro R, Bhatia KP, Hallett M. Refining the clinical diagnosis of Parkinson's disease. Parkinsonism Relat Disord 2024; 122:106041. [PMID: 38360507 PMCID: PMC11069446 DOI: 10.1016/j.parkreldis.2024.106041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Our ability to define, understand, and classify Parkinson's disease (PD) has undergone significant changes since the disorder was first described in 1817. Clinical features and neuropathologic signatures can now be supplemented by in-vivo interrogation of genetic and biological substrates of disease, offering great opportunity for further refining the diagnosis of PD. In this mini-review, we discuss the historical perspectives which shaped our thinking surrounding the definition and diagnosis of PD. We highlight the clinical, genetic, pathologic and biologic diversity which underpins the condition, and proceed to discuss how recent developments in our ability to define biologic and pathologic substrates of disease might impact PD definition, diagnosis, individualised prognostication, and personalised clinical care. We argue that Parkinson's 'disease', as currently diagnosed in the clinic, is actually a syndrome. It is the outward manifestation of any array of potential dysfunctional biologic processes, neuropathological changes, and disease aetiologies, which culminate in common outward clinical features which we term PD; each person has their own unique disease, which we can now define with increasing precision. This is an exciting time in PD research and clinical care. Our ability to refine the clinical diagnosis of PD, incorporating in-vivo assessments of disease biology, neuropathology, and neurogenetics may well herald the era of biologically-based, precision medicine approaches PD management. With this however comes a number of challenges, including how to integrate these technologies into clinical practice in a way which is acceptable to patients, promotes meaningful changes to care, and minimises health economic impact.
Collapse
Affiliation(s)
- Eoin Mulroy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, (SA), Italy
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
56
|
Hatano T, Okuzumi A, Matsumoto G, Tsunemi T, Hattori N. α-Synuclein: A Promising Biomarker for Parkinson's Disease and Related Disorders. J Mov Disord 2024; 17:127-137. [PMID: 38589016 PMCID: PMC11082597 DOI: 10.14802/jmd.24075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 03/30/2024] [Accepted: 04/09/2024] [Indexed: 04/10/2024] Open
Abstract
Mutations in the SNCA gene, which encodes α-synuclein (α-syn), play a key role in the development of genetic Parkinson's disease (PD). α-Syn is a major component of Lewy bodies in PD and glial cytoplasmic inclusions in multiple system atrophy (MSA). Rapid eye movement sleep behavior disorder patients often progress to PD, dementia with Lewy bodies, or MSA, which are collectively known as α-synucleinopathies. The loss of dopaminergic neurons with Lewy bodies precedes motor dysfunction in these diseases, but the mechanisms of neurodegeneration due to α-syn aggregation are poorly understood. Monitoring α-syn aggregation in vivo could serve as a diagnostic biomarker and help elucidate pathogenesis, necessitating a simple and accurate detection method. Seed amplification assays (SAAs), such as real-time quaking-induced conversion and protein misfolding cyclic amplification, are used to detect small amounts of abnormally structured α-syn protofibrils, which are central to aggregation. These methods are promising for the early diagnosis of α-synucleinopathy. Differences in α-syn filament structures between α-synucleinopathies, as observed through transmission electron microscopy and cryo-electron microscopy, suggest their role in the pathogenesis of neurodegeneration. SAAs may differentiate between subtypes of α-synucleinopathy and other diseases. Efforts are also being made to identify α-syn from blood using various methods. This review introduces body fluid α-syn biomarkers based on pathogenic α-syn seeds, which are expected to redefine α-synucleinopathy diagnosis and staging, improving clinical research accuracy and facilitating biomarker development.
Collapse
Affiliation(s)
- Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Gen Matsumoto
- Department of Histology and Cell Biology, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Taiji Tsunemi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
57
|
Painous C, Fernández M, Pérez J, de Mena L, Cámara A, Compta Y. Fluid and tissue biomarkers in Parkinson's disease: Immunodetection or seed amplification? Central or peripheral? Parkinsonism Relat Disord 2024; 121:105968. [PMID: 38168618 DOI: 10.1016/j.parkreldis.2023.105968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Over the last two decades there have been meaningful developments on biomarkers of neurodegenerative diseases, extensively (but not solely) focusing on their proteinopathic nature. Accordingly, in Alzheimer's disease determination of levels of total and phosphorylated tau (τ and p-τ, usually p-τ181) along with amyloid-beta1-42 (Aβ1-42) by immunodetection in cerebrospinal fluid (CSF) and currently even in peripheral blood, have been widely accepted and introduced to routine diagnosis. In the case of Parkinson's disease, α-synuclein as a potential biomarker (both for diagnosis and progression tracking) has proved more elusive under the immunodetection approach. In recent years, the emergence of the so-called seed amplification assays is proving to be a game-changer, with mounting evidence under different technical approaches and using a variety of biofluids or tissues, yielding promising diagnostic accuracies. Currently the least invasive but at once more reliable source of biosamples and techniques are being sought. Here we overview these advances.
Collapse
Affiliation(s)
- Celia Painous
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jesica Pérez
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Lorena de Mena
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ana Cámara
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
58
|
Hutchison RM, Fraser K, Yang M, Fox T, Hirschhorn E, Njingti E, Scott D, Bedell BJ, Kistner KM, Cedarbaum JM, Evans KC, Graham D, Martarello L, Mollenhauer B, Lang AE, Dam T, Beaver J. Cinpanemab in Early Parkinson Disease: Evaluation of Biomarker Results From the Phase 2 SPARK Clinical Trial. Neurology 2024; 102:e209137. [PMID: 38315945 DOI: 10.1212/wnl.0000000000209137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/29/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Sensitive, reliable, and scalable biomarkers are needed to accelerate the development of therapies for Parkinson disease (PD). In this study, we evaluate the biomarkers of early PD diagnosis, disease progression, and treatment effect collected in the SPARK. METHODS Cinpanemab is a human-derived monoclonal antibody binding preferentially to aggregated forms of extracellular α-synuclein. SPARK was a randomized, double-blind, placebo-controlled, phase 2 multicenter trial evaluating 3 cinpanemab doses administered intravenously every 4 weeks for 52 weeks with an active treatment dose-blind extension period for up to 112 weeks. SPARK enrolled 357 participants diagnosed with PD within 3 years, aged 40-80 years, ≤2.5 on the modified Hoehn and Yahr scale, and with evidence of striatal dopaminergic deficit. The primary outcome was change from baseline in the Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale total score. Secondary and exploratory biomarker outcomes evaluated change from baseline at week 52 relative to placebo. Dopamine transporter SPECT and MRI were used to quantify changes in the nigrostriatal dopamine pathway and regional atrophy. CSF and plasma samples were used to assess change in total α-synuclein levels, α-synuclein seeding, and neurofilament light chain levels. SPARK was conducted from January 2018 to April 2021 and terminated due to lack of efficacy. RESULTS Approximately 3.8% (15/398) of SPECT-imaged participants did not have evidence of dopaminergic deficit and were screen-failed. Binary classification of α-synuclein seeding designated 93% (110/118) of the enrolled CSF subgroup as positive for α-synuclein seeds at baseline. Clinical disease progression was observed, with no statistically significant difference in cinpanemab groups compared with that in placebo. Ninety-nine percent of participants with positive α-synuclein seeding remained positive through week 52. No statistically significant changes from baseline were observed between treatment groups and placebo across biomarker measures. Broadly, there was minimal annual change with high interindividual variability across biomarkers-with striatal binding ratios of the ipsilateral putamen showing the greatest mean change/SD over time. DISCUSSION Biomarker results indicated enrollment of the intended population with early PD, but there was no significant correlation with disease progression or clear evidence of a cinpanemab treatment effect on biomarker measures. Suitable biomarkers for evaluating disease severity and progression in early PD trials are still needed. TRIAL REGISTRATION INFORMATION NCT03318523 (clinicaltrials.gov/ct2/show/NCT03318523); Submitted October 24, 2017; First patient enrolled January 2018.
Collapse
Affiliation(s)
- R Matthew Hutchison
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Kyle Fraser
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Minhua Yang
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Tara Fox
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Elizabeth Hirschhorn
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Edwin Njingti
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - David Scott
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Barry J Bedell
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Kristi M Kistner
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Jesse M Cedarbaum
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Karleyton C Evans
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Danielle Graham
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Laurent Martarello
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Brit Mollenhauer
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Anthony E Lang
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Tien Dam
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - John Beaver
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| |
Collapse
|
59
|
Parnetti L, Paoletti FP. Fluid Biomarkers for Trial Enrichment and Effect Monitoring in Disease-Modifying Treatments for Parkinson Disease. Neurology 2024; 102:e209194. [PMID: 38315967 DOI: 10.1212/wnl.0000000000209194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024] Open
Affiliation(s)
- Lucilla Parnetti
- From the Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Italy
| | - Federico Paolini Paoletti
- From the Section of Neurology, Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Italy
| |
Collapse
|
60
|
Rutledge J, Lehallier B, Zarifkar P, Losada PM, Shahid-Besanti M, Western D, Gorijala P, Ryman S, Yutsis M, Deutsch GK, Mormino E, Trelle A, Wagner AD, Kerchner GA, Tian L, Cruchaga C, Henderson VW, Montine TJ, Borghammer P, Wyss-Coray T, Poston KL. Comprehensive proteomics of CSF, plasma, and urine identify DDC and other biomarkers of early Parkinson's disease. Acta Neuropathol 2024; 147:52. [PMID: 38467937 PMCID: PMC10927779 DOI: 10.1007/s00401-024-02706-0] [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: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024]
Abstract
Parkinson's disease (PD) starts at the molecular and cellular level long before motor symptoms appear, yet there are no early-stage molecular biomarkers for diagnosis, prognosis prediction, or monitoring therapeutic response. This lack of biomarkers greatly impedes patient care and translational research-L-DOPA remains the standard of care more than 50 years after its introduction. Here, we performed a large-scale, multi-tissue, and multi-platform proteomics study to identify new biomarkers for early diagnosis and disease monitoring in PD. We analyzed 4877 cerebrospinal fluid, blood plasma, and urine samples from participants across seven cohorts using three orthogonal proteomics methods: Olink proximity extension assay, SomaScan aptamer precipitation assay, and liquid chromatography-mass spectrometry proteomics. We discovered that hundreds of proteins were upregulated in the CSF, blood, or urine of PD patients, prodromal PD patients with DAT deficit and REM sleep behavior disorder or anosmia, and non-manifesting genetic carriers of LRRK2 and GBA mutations. We nominate multiple novel hits across our analyses as promising markers of early PD, including DOPA decarboxylase (DDC), also known as L-aromatic acid decarboxylase (AADC), sulfatase-modifying factor 1 (SUMF1), dipeptidyl peptidase 2/7 (DPP7), glutamyl aminopeptidase (ENPEP), WAP four-disulfide core domain 2 (WFDC2), and others. DDC, which catalyzes the final step in dopamine synthesis, particularly stands out as a novel hit with a compelling mechanistic link to PD pathogenesis. DDC is consistently upregulated in the CSF and urine of treatment-naïve PD, prodromal PD, and GBA or LRRK2 carrier participants by all three proteomics methods. We show that CSF DDC levels correlate with clinical symptom severity in treatment-naïve PD patients and can be used to accurately diagnose PD and prodromal PD. This suggests that urine and CSF DDC could be a promising diagnostic and prognostic marker with utility in both clinical care and translational research.
Collapse
Affiliation(s)
- Jarod Rutledge
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Pardis Zarifkar
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | - Patricia Moran Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Marian Shahid-Besanti
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Dan Western
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sephira Ryman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Translational Neuroscience, Mind Research Network, Albuquerque, NM, USA
| | - Maya Yutsis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Gayle K Deutsch
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Elizabeth Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Alexandra Trelle
- Department of Psychology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Anthony D Wagner
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Psychology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Geoffrey A Kerchner
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Roche Medical, Basel, Switzerland
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University School of Humanities and Sciences, Stanford University, Stanford, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- The Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- The Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
61
|
Vandendriessche C, Bruggeman A, Foroozandeh J, Van Hoecke L, Dujardin P, Xie J, Van Imschoot G, Van Wonterghem E, Castelein J, Lucci C, De Groef L, Vandenbroucke RE. The Spreading and Effects of Human Recombinant α-Synuclein Preformed Fibrils in the Cerebrospinal Fluid of Mice. eNeuro 2024; 11:ENEURO.0024-23.2024. [PMID: 38383588 PMCID: PMC10925901 DOI: 10.1523/eneuro.0024-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Parkinson's disease (PD) patients harbor seeding-competent α-synuclein (α-syn) in their cerebrospinal fluid (CSF), which is mainly produced by the choroid plexus (ChP). Nonetheless, little is known about the role of the CSF and the ChP in PD pathogenesis. To address this question, we used an intracerebroventricular (icv) injection mouse model to assess CSF α-syn spreading and its short- and long-term consequences on the brain. Hereby, we made use of seeding-competent, recombinant α-syn preformed fibrils (PFF) that are known to induce aggregation and subsequent spreading of endogenous α-syn in stereotactic tissue injection models. Here, we show that icv-injected PFF, but not monomers (Mono), are rapidly removed from the CSF by interaction with the ChP. Additionally, shortly after icv injection both Mono and PFF were detected in the olfactory bulb and striatum. This spreading was associated with increased inflammation and complement activation in these tissues as well as leakage of the blood-CSF barrier. Despite these effects, a single icv injection of PFF didn't induce a decline in motor function. In contrast, daily icv injections over the course of 5 days resulted in deteriorated grip strength and formation of phosphorylated α-syn inclusions in the brain 2 months later, whereas dopaminergic neuron levels were not affected. These results point toward an important clearance function of the CSF and the ChP, which could mediate removal of PFF from the brain, whereby chronic exposure to PFF in the CSF may negatively impact blood-CSF barrier functionality and PD pathology.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
- Department of Neurology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Joyce Foroozandeh
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
- VIB Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium
- Department of Neurosciences, Brain Institute KU Leuven, 3000, Leuven, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Junhua Xie
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Jonas Castelein
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Cristiano Lucci
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9000, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| |
Collapse
|
62
|
Huang J, Yuan X, Chen L, Hu B, Wang H, Wang Y, Huang W. Pathological α-synuclein detected by real-time quaking-induced conversion in synucleinopathies. Exp Gerontol 2024; 187:112366. [PMID: 38280659 DOI: 10.1016/j.exger.2024.112366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/10/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
synucleinopathies are diseases characterized by the aggregation of α-synuclein (α-syn), which forms fibrils through misfolding and accumulates in a prion-like manner. To detect the presence of these α-syn aggregates in clinical samples, seed amplification assays (SAAs) have been developed. These SAAs are capable of amplifying the α-syn seeds, allowing for their detection. αSyn-SAAs have been reported under the names 'protein misfolding cyclic amplification' (αSyn-PMCA) and 'real-time quaking-induced conversion'α-Syn-RT-QuIC. The α-Syn RT-QuIC, in particular, has been adapted to amplify and detect α-syn aggregates in various biospecimens, including cerebrospinal fluid (CSF), skin, nasal brushing, serum and saliva. The α-syn RT-QuIC assay has demonstrated good sensitivity and specificity in detecting pathological α-syn, particularly in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) cases, with an accuracy rate of up to 80 %. Additionally, differential diagnosis between DLB and PD, as well as PD and multiple system atrophy (MSA), can be achieved by utilizing certain kinetic thioflavin T (ThT) parameters and other parameters. Moreover, the positive detection of α-syn in the prodromal stage of synucleinopathies provides an opportunity for early intervention and management. In summary, the development of the α-syn RT-QuIC assay has greatly contributed to the field of synucleinopathies. Therefore, we review the development of α-syn RT-QuIC assay and describe in detail the recent advancements of α-syn RT-QuIC assay for detecting pathological α-syn in synucleinopathies.
Collapse
Affiliation(s)
- Juan Huang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Xingxing Yuan
- Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, China
| | - Lin Chen
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Binbin Hu
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Hui Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Ye Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China.
| | - Wei Huang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
63
|
Vaughan DP, Fumi R, Theilmann Jensen M, Georgiades T, Wu L, Lux D, Obrocki R, Lamoureux J, Ansorge O, Allinson K, Warner TT, Jaunmuktane Z, Misbahuddin A, Leigh PN, Ghosh B, Bhatia KP, Church A, Kobylecki C, Hu M, Rowe JB, Blauwendraat C, Morris HR, Jabbari E. Evaluation of cerebrospinal fluid alpha-synuclein seed amplification assay in PSP and CBS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.28.24303478. [PMID: 38529496 PMCID: PMC10962751 DOI: 10.1101/2024.02.28.24303478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Background Seed amplification assay (SAA) testing has become an important biomarker in the diagnosis of alpha-synuclein related neurodegenerative disorders. Objectives To assess the rate of alpha-synuclein SAA positivity in progressive supranuclear palsy (PSP) and corticobasal syndrome (CBS), and analyse the clinical and pathological features of SAA positive and negative cases. Methods 106 CSF samples from clinically diagnosed PSP (n=59), CBS (n=37) and indeterminate parkinsonism cases (n=10) were analysed using alpha-synuclein SAA. Results Three cases (1 PSP, 2 CBS) were Multiple System Atrophy (MSA)-type SAA positive. 5/59 (8.5%) PSP cases were Parkinson's disease (PD)-type SAA positive, and these cases were older and had a shorter disease duration compared with SAA negative cases. In contrast, 9/35 (25.7%) CBS cases were PD-type SAA positive. Conclusions Our results suggest that PD-type seeds can be detected in PSP and CBS using a CSF alpha-synuclein SAA, and in PSP this may impact on clinical course.
Collapse
|
64
|
Massey RS, Appadurai RR, Prakash R. A Surface Imprinted Polymer EIS Sensor for Detecting Alpha-Synuclein, a Parkinson's Disease Biomarker. MICROMACHINES 2024; 15:273. [PMID: 38399001 PMCID: PMC10892569 DOI: 10.3390/mi15020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Parkinson's Disease (PD) is a debilitating neurodegenerative disease, causing loss of motor function and, in some instances, cognitive decline and dementia in those affected. The quality of life can be improved, and disease progression delayed through early interventions. However, current methods of confirming a PD diagnosis are extremely invasive. This prevents their use as a screening tool for the early onset stages of PD. We propose a surface imprinted polymer (SIP) electroimpedance spectroscopy (EIS) biosensor for detecting α-Synuclein (αSyn) and its aggregates, a biomarker that appears in saliva and blood during the early stages of PD as the blood-brain barrier degrades. The surface imprinted polymer stamp is fabricated by low-temperature melt stamping polycaprolactone (PCL) on interdigitated EIS electrodes. The result is a low-cost, small-footprint biosensor that is highly suitable for non-invasive monitoring of the disease biomarker. The sensors were tested with αSyn dilutions in deionized water and in constant ionic concentration matrix solutions with decreasing concentrations of αSyn to remove the background effects of concentration. The device response confirmed the specificity of these devices to the target protein of monomeric αSyn. The sensor limit of detection was measured to be 5 pg/L, and its linear detection range was 5 pg/L-5 µg/L. This covers the physiological range of αSyn in saliva and makes this a highly promising method of quantifying αSyn monomers for PD patients in the future. The SIP surface was regenerated, and the sensor was reused to demonstrate its capability for repeat sensing as a potential continuous monitoring tool for the disease biomarker.
Collapse
Affiliation(s)
| | | | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; (R.S.M.); (R.R.A.)
| |
Collapse
|
65
|
Bartz JC, Benavente R, Caughey B, Christensen S, Herbst A, Hoover EA, Mathiason CK, McKenzie D, Morales R, Schwabenlander MD, Walsh DP. Chronic Wasting Disease: State of the Science. Pathogens 2024; 13:138. [PMID: 38392876 PMCID: PMC10892334 DOI: 10.3390/pathogens13020138] [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: 12/18/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic wasting disease (CWD) is a prion disease affecting cervid species, both free-ranging and captive populations. As the geographic range continues to expand and disease prevalence continues to increase, CWD will have an impact on cervid populations, local economies, and ecosystem health. Mitigation of this "wicked" disease will require input from many different stakeholders including hunters, landowners, research biologists, wildlife managers, and others, working together. The NC1209 (North American interdisciplinary chronic wasting disease research consortium) is composed of scientists from different disciplines involved with investigating and managing CWD. Leveraging this broad breadth of expertise, the Consortium has created a state-of-the-science review of five key aspects of CWD, including current diagnostic capabilities for detecting prions, requirements for validating these diagnostics, the role of environmental transmission in CWD dynamics, and potential zoonotic risks associated with CWD. The goal of this review is to increase stakeholders', managers', and decision-makers' understanding of this disease informed by current scientific knowledge.
Collapse
Affiliation(s)
- Jason C. Bartz
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, NE 68178, USA;
| | - Rebeca Benavente
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (R.B.); (R.M.)
| | - Byron Caughey
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Sonja Christensen
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48824, USA;
| | - Allen Herbst
- U.S. Geological Survey, National Wildlife Health Center, Madison, WI 53711, USA;
| | - Edward A. Hoover
- Prion Research Center, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (E.A.H.); (C.K.M.)
| | - Candace K. Mathiason
- Prion Research Center, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (E.A.H.); (C.K.M.)
| | - Debbie McKenzie
- Department of Biological Sciences, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M9, Canada;
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (R.B.); (R.M.)
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Marc D. Schwabenlander
- Minnesota Center for Prion Research and Outreach, Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Daniel P. Walsh
- U.S. Geological Survey, Montana Cooperative Wildlife Research Unit, University of Montana, Missoula, MT 59812, USA
| | | |
Collapse
|
66
|
Ndayisaba A, Pitaro AT, Willett AS, Jones KA, de Gusmao CM, Olsen AL, Kim J, Rissanen E, Woods JK, Srinivasan SR, Nagy A, Nagy A, Mesidor M, Cicero S, Patel V, Oakley DH, Tuncali I, Taglieri-Noble K, Clark EC, Paulson J, Krolewski RC, Ho GP, Hung AY, Wills AM, Hayes MT, Macmore JP, Warren L, Bower PG, Langer CB, Kellerman LR, Humphreys CW, Glanz BI, Dielubanza EJ, Frosch MP, Freeman RL, Gibbons CH, Stefanova N, Chitnis T, Weiner HL, Scherzer CR, Scholz SW, Vuzman D, Cox LM, Wenning G, Schmahmann JD, Gupta AS, Novak P, Young GS, Feany MB, Singhal T, Khurana V. Clinical Trial-Ready Patient Cohorts for Multiple System Atrophy: Coupling Biospecimen and iPSC Banking to Longitudinal Deep-Phenotyping. CEREBELLUM (LONDON, ENGLAND) 2024; 23:31-51. [PMID: 36190676 PMCID: PMC9527378 DOI: 10.1007/s12311-022-01471-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
Multiple system atrophy (MSA) is a fatal neurodegenerative disease of unknown etiology characterized by widespread aggregation of the protein alpha-synuclein in neurons and glia. Its orphan status, biological relationship to Parkinson's disease (PD), and rapid progression have sparked interest in drug development. One significant obstacle to therapeutics is disease heterogeneity. Here, we share our process of developing a clinical trial-ready cohort of MSA patients (69 patients in 2 years) within an outpatient clinical setting, and recruiting 20 of these patients into a longitudinal "n-of-few" clinical trial paradigm. First, we deeply phenotype our patients with clinical scales (UMSARS, BARS, MoCA, NMSS, and UPSIT) and tests designed to establish early differential diagnosis (including volumetric MRI, FDG-PET, MIBG scan, polysomnography, genetic testing, autonomic function tests, skin biopsy) or disease activity (PBR06-TSPO). Second, we longitudinally collect biospecimens (blood, CSF, stool) and clinical, biometric, and imaging data to generate antecedent disease-progression scores. Third, in our Mass General Brigham SCiN study (stem cells in neurodegeneration), we generate induced pluripotent stem cell (iPSC) models from our patients, matched to biospecimens, including postmortem brain. We present 38 iPSC lines derived from MSA patients and relevant disease controls (spinocerebellar ataxia and PD, including alpha-synuclein triplication cases), 22 matched to whole-genome sequenced postmortem brain. iPSC models may facilitate matching patients to appropriate therapies, particularly in heterogeneous diseases for which patient-specific biology may elude animal models. We anticipate that deeply phenotyped and genotyped patient cohorts matched to cellular models will increase the likelihood of success in clinical trials for MSA.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Ariana T Pitaro
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Andrew S Willett
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Kristie A Jones
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Claudio Melo de Gusmao
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Abby L Olsen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jisoo Kim
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Eero Rissanen
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jared K Woods
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sharan R Srinivasan
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI , 48103, USA
| | - Anna Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Amanda Nagy
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Merlyne Mesidor
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Steven Cicero
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Viharkumar Patel
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Idil Tuncali
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Katherine Taglieri-Noble
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Emily C Clark
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jordan Paulson
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Richard C Krolewski
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gary P Ho
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Albert Y Hung
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anne-Marie Wills
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael T Hayes
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Jason P Macmore
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | | | - Pamela G Bower
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Carol B Langer
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Lawrence R Kellerman
- The Multiple System Atrophy Coalition, Inc., 7918 Jones Branch Drive, Suite 300, McLean, VA, 22102, USA
| | - Christopher W Humphreys
- Department of Pulmonary, Sleep and Critical Care Medicine, Salem Hospital, MassGeneral Brigham, Salem, MA, 01970, USA
| | - Bonnie I Glanz
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Elodi J Dielubanza
- Department of Urology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Roy L Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Christopher H Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Nadia Stefanova
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Tanuja Chitnis
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Howard L Weiner
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Clemens R Scherzer
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Sonja W Scholz
- Laboratory of Neurogenetics, Disorders and Stroke, National Institute of Neurological, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, 21287, USA
| | - Dana Vuzman
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Laura M Cox
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Gregor Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anoopum S Gupta
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Peter Novak
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Geoffrey S Young
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Tarun Singhal
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA
| | - Vikram Khurana
- Department of Neurology, Building for Transformative Medicine Room 10016L, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, 02115, USA.
| |
Collapse
|
67
|
Höglinger GU, Adler CH, Berg D, Klein C, Outeiro TF, Poewe W, Postuma R, Stoessl AJ, Lang AE. A biological classification of Parkinson's disease: the SynNeurGe research diagnostic criteria. Lancet Neurol 2024; 23:191-204. [PMID: 38267191 DOI: 10.1016/s1474-4422(23)00404-0] [Citation(s) in RCA: 188] [Impact Index Per Article: 188.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/08/2023] [Accepted: 10/06/2023] [Indexed: 01/26/2024]
Abstract
With the hope that disease-modifying treatments could target the molecular basis of Parkinson's disease, even before the onset of symptoms, we propose a biologically based classification. Our classification acknowledges the complexity and heterogeneity of the disease by use of a three-component system (SynNeurGe): presence or absence of pathological α-synuclein (S) in tissues or CSF; evidence of underlying neurodegeneration (N) defined by neuroimaging procedures; and documentation of pathogenic gene variants (G) that cause or strongly predispose to Parkinson's disease. These three components are linked to a clinical component (C), defined either by a single high-specificity clinical feature or by multiple lower-specificity clinical features. The use of a biological classification will enable advances in both basic and clinical research, and move the field closer to the precision medicine required to develop disease-modifying therapies. We emphasise the initial application of these criteria exclusively for research. We acknowledge its ethical implications, its limitations, and the need for prospective validation in future studies.
Collapse
Affiliation(s)
- Günter U Höglinger
- Department of Neurology, University Hospital, Ludwig-Maximilians-University (LMU) and German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Daniela Berg
- Christian Albrechts University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lüebeck, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Werner Poewe
- Medical University Innsbruck, Innsbruck, Austria
| | - Ronald Postuma
- Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, QC, Canada
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and Parkinson's Foundation Centre of Excellence, University of British Columbia, BC, Canada
| | - Anthony E Lang
- University Health Network's Krembil Brain Institute, Edmond J Safra Program in Parkinson's Disease and the Rossy PSP Centre, Toronto Western Hospital, Toronto, ON, Canada.
| |
Collapse
|
68
|
Cardoso F, Goetz CG, Mestre TA, Sampaio C, Adler CH, Berg D, Bloem BR, Burn DJ, Fitts MS, Gasser T, Klein C, de Tijssen MAJ, Lang AE, Lim SY, Litvan I, Meissner WG, Mollenhauer B, Okubadejo N, Okun MS, Postuma RB, Svenningsson P, Tan LCS, Tsunemi T, Wahlstrom-Helgren S, Gershanik OS, Fung VSC, Trenkwalder C. A Statement of the MDS on Biological Definition, Staging, and Classification of Parkinson's Disease. Mov Disord 2024; 39:259-266. [PMID: 38093469 DOI: 10.1002/mds.29683] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Affiliation(s)
- Francisco Cardoso
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, The Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Christopher G Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Tiago A Mestre
- Ottawa Hospital Research Institute; University of Ottawa Brain and Mind Research Institute; Division of Neurology, Department of Medicine, University of Ottawa, The Ottawa Hospital Ottawa, Ottawa, Ontario, Canada
| | - Cristina Sampaio
- CHDI Management/CHDI Foundation, Princeton, New Jersey, USA
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Christian Albrechts-University of Kiel, Kiel, Germany
| | - Bastiaan R Bloem
- Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Department of Neurology, Center of Expertise for Parkinson and Movement Disorders, Nijmegen, The Netherlands
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael S Fitts
- UAB Libraries, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Thomas Gasser
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany. German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Marina A J de Tijssen
- Department of Neurology, Expertise Centre Movement Disorders, University Medical Centre Groningen, Groningen, The Netherlands
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, University Health Network and the University of Toronto, Toronto, Ontario, Canada
| | - Shen-Yang Lim
- Division of Neurology, Department of Medicine, and the Mah Pooi Soo and Tan Chin Nam Centre for Parkinson's and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Wassilios G Meissner
- CHU Bordeaux, Service de Neurologie des Maladies Neurodégénératives, Bordeaux, France
- Univ. Bordeaux, CNRS, IMN, Bordeaux, France
- Department of Medicine, University of Otago, Christchurch, and New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center, Kassel, Germany
| | - Njideka Okubadejo
- Neurology Unit, Department of Medicine, Faculty of Clinical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Michael S Okun
- Adelaide Lackner Professor of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainsville, Florida, USA
| | - Ronald B Postuma
- Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | | | | | - Taiji Tsunemi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Oscar S Gershanik
- Movement Disorders Unit, Institute of Neuroscience, Favaloro Foundation University Hospital, Buenos Aires, Argentina
- Cognitive Neuroscience Laboratory, Institute of Cognitive Neurology (INECO), Buenos Aires, Argentina
| | - Victor S C Fung
- Movement Disorders Unit, Department of Neurology, Westmead Hospital and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Claudia Trenkwalder
- Paracelsus-Elena Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center, Goettingen, Germany
| |
Collapse
|
69
|
Simuni T, Chahine LM, Poston K, Brumm M, Buracchio T, Campbell M, Chowdhury S, Coffey C, Concha-Marambio L, Dam T, DiBiaso P, Foroud T, Frasier M, Gochanour C, Jennings D, Kieburtz K, Kopil CM, Merchant K, Mollenhauer B, Montine T, Nudelman K, Pagano G, Seibyl J, Sherer T, Singleton A, Stephenson D, Stern M, Soto C, Tanner CM, Tolosa E, Weintraub D, Xiao Y, Siderowf A, Dunn B, Marek K. A biological definition of neuronal α-synuclein disease: towards an integrated staging system for research. Lancet Neurol 2024; 23:178-190. [PMID: 38267190 DOI: 10.1016/s1474-4422(23)00405-2] [Citation(s) in RCA: 208] [Impact Index Per Article: 208.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 01/26/2024]
Abstract
Parkinson's disease and dementia with Lewy bodies are currently defined by their clinical features, with α-synuclein pathology as the gold standard to establish the definitive diagnosis. We propose that, given biomarker advances enabling accurate detection of pathological α-synuclein (ie, misfolded and aggregated) in CSF using the seed amplification assay, it is time to redefine Parkinson's disease and dementia with Lewy bodies as neuronal α-synuclein disease rather than as clinical syndromes. This major shift from a clinical to a biological definition of Parkinson's disease and dementia with Lewy bodies takes advantage of the availability of tools to assess the gold standard for diagnosis of neuronal α-synuclein (n-αsyn) in human beings during life. Neuronal α-synuclein disease is defined by the presence of pathological n-αsyn species detected in vivo (S; the first biological anchor) regardless of the presence of any specific clinical syndrome. On the basis of this definition, we propose that individuals with pathological n-αsyn aggregates are at risk for dopaminergic neuronal dysfunction (D; the second biological anchor). Our biological definition establishes a staging system, the neuronal α-synuclein disease integrated staging system (NSD-ISS), rooted in the biological anchors (S and D) and the degree of functional impairment caused by clinical signs or symptoms. Stages 0-1 occur without signs or symptoms and are defined by the presence of pathogenic variants in the SNCA gene (stage 0), S alone (stage 1A), or S and D (stage 1B). The presence of clinical manifestations marks the transition to stage 2 and beyond. Stage 2 is characterised by subtle signs or symptoms but without functional impairment. Stages 2B-6 require both S and D and stage-specific increases in functional impairment. A biological definition of neuronal α-synuclein disease and an NSD-ISS research framework are essential to enable interventional trials at early disease stages. The NSD-ISS will evolve to include the incorporation of data-driven definitions of stage-specific functional anchors and additional biomarkers as they emerge and are validated. Presently, the NSD-ISS is intended for research use only; its application in the clinical setting is premature and inappropriate.
Collapse
Affiliation(s)
- Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathleen Poston
- Department of Neurology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Teresa Buracchio
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Michelle Campbell
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sohini Chowdhury
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | | | | | - Peter DiBiaso
- Patient Advisory Council, New York, NY, USA; Clinical Solutions and Strategic Partnerships, WCG Clinical, Princeton, NJ, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Mark Frasier
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Caroline Gochanour
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | | | - Karl Kieburtz
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Catherine M Kopil
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen and Paracelsus-Elena-Klinik, Kassel, Germany
| | - Thomas Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kelly Nudelman
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | | | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Todd Sherer
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Andrew Singleton
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Diane Stephenson
- Critical Path for Parkinson's, Critical Path Institute, Tucson, AZ, USA
| | - Matthew Stern
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudio Soto
- Amprion, San Diego, CA, USA; Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Caroline M Tanner
- Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, CA, USA; Parkinson's Disease Research Education and Clinical Center, San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
| | - Eduardo Tolosa
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Hospital Clínic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Daniel Weintraub
- University of Pennsylvania and the Parkinson's Disease and Mental Illness Research, Education and Clinical Centers, Philadelphia Veterans Affairs Medical Center Philadelphia, PA, USA
| | - Yuge Xiao
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Billy Dunn
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| |
Collapse
|
70
|
Brockmann K, Lerche S, Baiardi S, Rossi M, Wurster I, Quadalti C, Roeben B, Mammana A, Zimmermann M, Hauser AK, Deuschle C, Schulte C, Liepelt-Scarfone I, Gasser T, Parchi P. CSF α-synuclein seed amplification kinetic profiles are associated with cognitive decline in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:24. [PMID: 38242875 PMCID: PMC10799016 DOI: 10.1038/s41531-023-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024] Open
Abstract
Seed amplification assays have been implemented in Parkinson's disease to reveal disease-specific misfolded alpha-synuclein aggregates in biospecimens. While the assays' qualitative dichotomous seeding response is valuable to stratify and enrich cohorts for alpha-synuclein pathology in general, more quantitative parameters that are associated with clinical dynamics of disease progression and that might potentially serve as exploratory outcome measures in clinical trials targeting alpha-synuclein would add important information. To evaluate whether the seeding kinetic parameters time required to reach the seeding threshold (LAG phase), the peak of fluorescence response (Imax), and the area under the curve (AUC) are associated with clinical trajectories, we analyzed LAG, Imax, and AUC in relation to the development of cognitive decline in a longitudinal cohort of 199 people with Parkinson's disease with positive CSF alpha-synuclein seeding status. Patients were stratified into tertiles based on their individual CSF alpha-synuclein seeding kinetic properties. The effect of the kinetic parameters on longitudinal development of cognitive impairment defined by MoCA ≤25 was analyzed by Cox-Regression. Patients with a higher number of positive seeding replicates and tertile groups of shorter LAG, higher Imax, and higher AUC showed a higher prevalence of and a shorter duration until cognitive impairment longitudinally (3, 6, and 3 years earlier with p ≤ 0.001, respectively). Results remained similar in separate subgroup analyses of patients with and without GBA mutation. We conclude that a more prominent alpha-synuclein seeding kinetic profile translates into a more rapid development of cognitive decline.
Collapse
Affiliation(s)
- Kathrin Brockmann
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany.
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany.
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
| | - Stefanie Lerche
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
| | - Isabel Wurster
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
- Edmond J. Safra Fellow in Movement Disorders, Tuebingen, Germany
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Benjamin Roeben
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
| | - Milan Zimmermann
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Ann-Kathrin Hauser
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Christian Deuschle
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Inga Liepelt-Scarfone
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Thomas Gasser
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
| |
Collapse
|
71
|
Bentivenga GM, Mammana A, Baiardi S, Rossi M, Ticca A, Magliocchetti F, Mastrangelo A, Poleggi A, Ladogana A, Capellari S, Parchi P. Performance of a seed amplification assay for misfolded alpha-synuclein in cerebrospinal fluid and brain tissue in relation to Lewy body disease stage and pathology burden. Acta Neuropathol 2024; 147:18. [PMID: 38240849 PMCID: PMC10799141 DOI: 10.1007/s00401-023-02663-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 01/22/2024]
Abstract
The development of in vitro seed amplification assays (SAA) detecting misfolded alpha-synuclein (αSyn) in cerebrospinal fluid (CSF) and other tissues has provided a pathology-specific biomarker for Lewy body disease (LBD). However, αSyn SAA diagnostic performance in early pathological stages or low Lewy body (LB) pathology load has only been assessed in small cohorts. Moreover, the relationship between SAA kinetic parameters, the number of αSyn brain seeds and the LB pathology burden assessed by immunohistochemistry has never been systematically investigated. We tested 269 antemortem CSF samples and 138 serially diluted brain homogenates from patients with and without neuropathological evidence of LBD in different stages by the αSyn Real-Time Quaking-Induced Conversion (RT-QuIC) SAA. Moreover, we looked for LB pathology by αSyn immunohistochemistry in a consecutive series of 604 Creutzfeldt-Jakob disease (CJD)-affected brains. αSyn CSF RT-QuIC showed 100% sensitivity in detecting LBD in limbic and neocortical stages. The assay sensitivity was significantly lower in patients in early stages (37.5% in Braak 1 and 2, 73.3% in Braak 3) or with focal pathology (50% in amygdala-predominant). The average number of CSF RT-QuIC positive replicates significantly correlated with LBD stage. Brain homogenate RT-QuIC showed higher sensitivity than immunohistochemistry for the detection of misfolded αSyn. In the latter, the kinetic parameter lag phase (time to reach the positive threshold) strongly correlated with the αSyn seed concentration in serial dilution experiments. Finally, incidental LBD prevalence was 8% in the CJD cohort. The present results indicate that (a) CSF RT-QuIC has high specificity and sufficient sensitivity to detect all patients with LB pathology at Braak stages > 3 and most of those at stage 3; (b) brain deposition of misfolded αSyn precedes the formation of LB and Lewy neurites; (c) αSyn SAA provides "quantitative" information regarding the LB pathology burden, with the lag phase and the number of positive replicates being the most promising variables to be used in the clinical setting.
Collapse
Affiliation(s)
| | - Angela Mammana
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marcello Rossi
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | - Andrea Mastrangelo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Poleggi
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Ladogana
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Sabina Capellari
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Piero Parchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy.
| |
Collapse
|
72
|
Gilboa T, Swank Z, Thakur R, Gould RA, Ooi KH, Norman M, Flynn EA, Deveney BT, Chen A, Borberg E, Kuzkina A, Ndayisaba A, Khurana V, Weitz DA, Walt DR. Toward the quantification of α-synuclein aggregates with digital seed amplification assays. Proc Natl Acad Sci U S A 2024; 121:e2312031121. [PMID: 38194461 PMCID: PMC10801878 DOI: 10.1073/pnas.2312031121] [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: 07/14/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024] Open
Abstract
The quantification and characterization of aggregated α-synuclein in clinical samples offer immense potential toward diagnosing, treating, and better understanding neurodegenerative synucleinopathies. Here, we developed digital seed amplification assays to detect single α-synuclein aggregates by partitioning the reaction into microcompartments. Using pre-formed α-synuclein fibrils as reaction seeds, we measured aggregate concentrations as low as 4 pg/mL. To improve our sensitivity, we captured aggregates on antibody-coated magnetic beads before running the amplification reaction. By first characterizing the pre-formed fibrils with transmission electron microscopy and size exclusion chromatography, we determined the specific aggregates targeted by each assay platform. Using brain tissue and cerebrospinal fluid samples collected from patients with Parkinson's Disease and multiple system atrophy, we demonstrated that the assay can detect endogenous pathological α-synuclein aggregates. Furthermore, as another application for these assays, we studied the inhibition of α-synuclein aggregation in the presence of small-molecule inhibitors and used a custom image analysis pipeline to quantify changes in aggregate growth and filament morphology.
Collapse
Affiliation(s)
- Tal Gilboa
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Harvard Medical School, Boston, MA02115
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Harvard Medical School, Boston, MA02115
| | - Rohan Thakur
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA02138
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Russell A. Gould
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Kean Hean Ooi
- Department of Medical Sciences, Harvard Medical School, Boston, MA02115
| | - Maia Norman
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Harvard Medical School, Boston, MA02115
- Physician Scientist Training Program, Massachusetts General Hospital/McLean Residency in Adult Psychiatry, Boston, MA02114
| | - Elizabeth A. Flynn
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Brendan T. Deveney
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA02138
| | - Anqi Chen
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA02138
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Harvard Medical School, Boston, MA02115
| | - Anastasia Kuzkina
- Harvard Medical School, Boston, MA02115
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA02115
| | - Alain Ndayisaba
- Harvard Medical School, Boston, MA02115
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA02115
| | - Vikram Khurana
- Harvard Medical School, Boston, MA02115
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Stem Cell Institute, Cambridge, MA02138
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - David A. Weitz
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Physics, Harvard University, Cambridge, MA02138
| | - David R. Walt
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Harvard Medical School, Boston, MA02115
| |
Collapse
|
73
|
Rojvirat C, Arismendi GR, Feinstein E, Guzman M, Citron BA, Delic V. Systematic Review of Post-Traumatic Parkinsonism, an Emerging Parkinsonian Disorder Among Survivors of Traumatic Brain Injury. Neurotrauma Rep 2024; 5:37-49. [PMID: 38292732 PMCID: PMC10825274 DOI: 10.1089/neur.2023.0104] [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] [Indexed: 02/01/2024] Open
Abstract
This systematic review focuses on an increasing subset of traumatic brain injury (TBI) survivors who develop post-traumatic parkinsonism (PTP), characterized by slowness of movement (bradykinesia), rigidity (stiffness), postural instability, and resting tremors caused by obstruction or damage to deep brain structures of the basal ganglia. PTP is rare, and one hypothesis to explain PTP rarity is that TBIs severe enough to affect deep brain structures are often lethal; however, with increasing survivability of TBIs, these numbers are expected to increase. The goal of this review is to raise awareness of an expected global increase of a subgroup of TBI patients who are treatment responsive and report therapeutic results aiding providers in diagnosing, educating, and treating PTP patients. Literature over the past 100 years was considered, and 44,663 peer-reviewed articles were identified. Inclusion criteria required a clinical indication of parkinsonian signs and TBI. Twenty-six case reports were ultimately included from which 36 individual patient data points were extracted for this review. Between 1980 and 2010, there has been an increase in reporting of PTP decade after decade. Forty-seven percent of PTP cases have 1-6 months of latency to symptom onset, and 83% of cases were male. PTP can occur with or without presence of brain lesions, and the most common type of injuries that cause PTP are motor vehicle accidents followed by falls. PTP patients are responsive to surgery or medication treatments. Further detail on PTP symptomology, treatment responsiveness, and injury types is provided.
Collapse
Affiliation(s)
- Catherine Rojvirat
- Laboratory of Molecular Biology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Neurology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Neurology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Gabriel R. Arismendi
- Laboratory of Molecular Biology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Neurology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Neurology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Erin Feinstein
- Department of Neurology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Maynard Guzman
- Laboratory of Molecular Biology, VA New Jersey Health Care System, East Orange, New Jersey, USA
| | - Bruce A. Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, East Orange, New Jersey, USA
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
74
|
Schaeffer E, Kluge A, Schulte C, Deuschle C, Bunk J, Welzel J, Maetzler W, Berg D. Association of Misfolded α-Synuclein Derived from Neuronal Exosomes in Blood with Parkinson's Disease Diagnosis and Duration. JOURNAL OF PARKINSON'S DISEASE 2024; 14:667-679. [PMID: 38669557 PMCID: PMC11191501 DOI: 10.3233/jpd-230390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 04/28/2024]
Abstract
Background Misfolded α-synuclein can be detected in blood samples of Parkinson's disease (PD) patients by a seed amplification assay (SAA), but the association with disease duration is not clear, yet. Objective In the present study we aimed to elucidate whether seeding activity of misfolded α-synuclein derived from neuronal exosomes in blood is associated with PD diagnosis and disease duration. Methods Cross-sectional samples of PD patients were analyzed and compared to samples of age- and gender-matched healthy controls using a blood-based SAA. Presence of α-synuclein seeding activity and differences in seeding parameters, including fluorescence response (in arbitrary units) at the end of the amplification assay (F60) were analyzed. Additionally, available PD samples collected longitudinally over 5-9 years were included. Results In the cross-sectional dataset, 79 of 80 PD patients (mean age 69 years, SD = 8; 56% male) and none of the healthy controls (n = 20, mean age 70 years, SD = 10; 55% male) showed seeding activity (sensitivity 98.8%). When comparing subgroups divided by disease duration, longer disease duration was associated with lower α-synuclein seeding activity (F60: p < 0.001). In the longitudinal analysis 10/11 patients showed a gradual decrease of α-synuclein seeding activity over time. Conclusions This study confirms the high sensitivity of the blood-based α-synuclein SAA applied here. The negative association of α-synuclein seeding activity in blood with disease duration makes this parameter potentially interesting as biomarker for future studies on the pathophysiology of disease progression in PD, and for biologically oriented trials in this field.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Annika Kluge
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Christian Deuschle
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Josina Bunk
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Julius Welzel
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| |
Collapse
|
75
|
Kluge A, Iranzo A. Biofluid Detection of Pathological α-Synuclein in the Prodromal Phase of Synucleinopathies. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S323-S331. [PMID: 38995801 PMCID: PMC11494638 DOI: 10.3233/jpd-230429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 07/14/2024]
Abstract
Synucleinopathies are disorders characterized by the aggregation and deposition of pathological α-synuclein conformers. The underlying neurodegenerative processes begin years or decades before the onset of cardinal motor symptoms. This prodromal phase may manifest with various signs or symptoms. However, there are no current standardized laboratory tests to ascertain the progression and conversion of prodromal conditions such as mild cognitive impairment, isolated REM sleep behavior disorder or pure autonomic failure. The aim of this systematic review was to evaluate the diagnostic possibilities using human biofluids as source material to detect pathological α-synuclein in the prodromal phase of synucleinopathies. Our review identified eight eligible studies, that investigated pathological α-synuclein conformers using cerebrospinal fluid from patients with prodromal signs of synulceinopathies to differentiate this patient group from non-synucleinopathies, while only one study investigated this aspect using blood as medium. While previous studies clearly demonstrated a high diagnostic performance of α-synuclein seed amplification assays for differentiating synucleinopathies with Lewy bodies from healthy controls, only few analyses were performed focussing on individuals with prodromal disease. Nevertheless, results for the early detection of α-synuclein seeds using α-synuclein seed amplification assays were promising and may be of particular relevance for future clinical trials and clinical practice.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Kiel University, Kiel, Germany
| | - Alex Iranzo
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, Barcelona University, IDIBAPS, CIBERNED, Barcelona, Spain
| |
Collapse
|
76
|
Schaeffer E, Yilmaz R, St. Louis EK, Noyce AJ. Ethical Considerations for Identifying Individuals in the Prodromal/Early Phase of Parkinson's Disease: A Narrative Review. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S307-S319. [PMID: 38995800 PMCID: PMC11492008 DOI: 10.3233/jpd-230428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
The ability to identify individuals in the prodromal phase of Parkinson's disease has improved in recent years, raising the question of whether and how those affected should be informed about the risk of future disease. Several studies investigated prognostic counselling for individuals with isolated REM sleep behavior disorder and have shown that most patients want to receive information about prognosis, but autonomy and individual preferences must be respected. However, there are still many unanswered questions about risk disclosure or early diagnosis of PD, including the impact on personal circumstances, cultural preferences and specific challenges associated with different profiles of prodromal symptoms, genetic testing or biomarker assessments. This narrative review aims to summarize the current literature on prognostic counselling and risk disclosure in PD, as well as highlight future perspectives that may emerge with the development of new biomarkers and their anticipated impact on the definition of PD.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Kiel University, Kiel, Germany
| | - Rezzak Yilmaz
- Department of Neurology, Ankara University School of Medicine, Ankara, Turkey
- Ankara University Brain Research Center, Ankara, Turkey
| | - Erik K. St. Louis
- Mayo Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, USA
| | - Alastair J. Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
77
|
Saramowicz K, Siwecka N, Galita G, Kucharska-Lusina A, Rozpędek-Kamińska W, Majsterek I. Alpha-Synuclein Contribution to Neuronal and Glial Damage in Parkinson's Disease. Int J Mol Sci 2023; 25:360. [PMID: 38203531 PMCID: PMC10778752 DOI: 10.3390/ijms25010360] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra and the widespread accumulation of alpha-synuclein (αSyn) protein aggregates. αSyn aggregation disrupts critical cellular processes, including synaptic function, mitochondrial integrity, and proteostasis, which culminate in neuronal cell death. Importantly, αSyn pathology extends beyond neurons-it also encompasses spreading throughout the neuronal environment and internalization by microglia and astrocytes. Once internalized, glia can act as neuroprotective scavengers, which limit the spread of αSyn. However, they can also become reactive, thereby contributing to neuroinflammation and the progression of PD. Recent advances in αSyn research have enabled the molecular diagnosis of PD and accelerated the development of targeted therapies. Nevertheless, despite more than two decades of research, the cellular function, aggregation mechanisms, and induction of cellular damage by αSyn remain incompletely understood. Unraveling the interplay between αSyn, neurons, and glia may provide insights into disease initiation and progression, which may bring us closer to exploring new effective therapeutic strategies. Herein, we provide an overview of recent studies emphasizing the multifaceted nature of αSyn and its impact on both neuron and glial cell damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (K.S.); (N.S.); (G.G.); (A.K.-L.); (W.R.-K.)
| |
Collapse
|
78
|
Zheng Y, Li S, Yang C, Yu Z, Jiang Y, Feng T. Comparison of biospecimens for α-synuclein seed amplification assays in Parkinson's disease: A systematic review and network meta-analysis. Eur J Neurol 2023; 30:3949-3967. [PMID: 37573472 DOI: 10.1111/ene.16041] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/23/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND AND PURPOSE Alpha-synuclein seed amplification assays (α-syn SAAs) are promising diagnostic methods for Parkinson's disease (PD) and other synucleinopathies. However, there is limited consensus regarding the diagnostic and differential diagnostic performance of α-syn SAAs on biofluids and peripheral tissues. METHODS A comprehensive research was performed in PubMed, Web of Science, Embase and Cochrane Library. Meta-analysis was performed using a random-effects model. A network meta-analysis based on an ANOVA model was conducted to compare the relative accuracy of α-syn SAAs with different specimens. RESULTS The pooled sensitivity and specificity of α-syn SAAs in distinguishing PD from healthy controls or non-neurodegenerative neurological controls were 0.91 (95% confidence interval [CI] 0.89-0.92) and 0.95 (95% CI 0.94-0.96) for cerebrospinal fluid (CSF); 0.91 (95% CI 0.86-0.94) and 0.92 (95% CI 0.87-0.95) for skin; 0.80 (95% CI 0.66-0.89) and 0.87 (95% CI 0.69-0.96) for submandibular gland; 0.44 (95% CI 0.30-0.59) and 0.92 (95% CI 0.79-0.98) for gastrointestinal tract; 0.79 (95% CI 0.70-0.86) and 0.88 (95% CI 0.77-0.95) for saliva; and 0.51 (95% CI 0.39-0.62) and 0.91 (95% CI 0.84-0.96) for olfactory mucosa (OM). The pooled sensitivity and specificity were 0.91 (95% CI 0.89-0.93) and 0.50 (95% CI 0.44-0.55) for CSF, 0.92 (95% CI 0.83-0.97) and 0.22 (95% CI 0.06-0.48) for skin, and 0.55 (95% CI 0.42-0.68) and 0.50 (95% CI 0.35-0.65) for OM in distinguishing PD from multiple system atrophy. The pooled sensitivity and specificity were 0.92 (95% CI 0.89-0.94) and 0.84 (95% CI 0.73-0.91) for CSF, 0.92 (95% CI 0.83-0.97) and 0.88 (95% CI 0.64-0.99) for skin and 0.63 (95% CI 0.52-0.73) and 0.86 (95% CI 0.64-0.97) for OM in distinguishing PD from progressive supranuclear palsy. The pooled sensitivity and specificity were 0.94 (95% CI 0.90-0.97) and 0.95 (95% CI 0.77-1.00) for CSF and 0.94 (95% CI 0.84-0.99) and 0.86 (95% CI 0.42-1.00) for skin in distinguishing PD from corticobasal degeneration. CONCLUSIONS α-Synuclein SAAs of CSF, skin, saliva, submandibular gland, gastrointestinal tract and OM are promising diagnostic assays for PD, with CSF and skin α-syn SAAs demonstrating higher diagnostic performance.
Collapse
Affiliation(s)
- Yuanchu Zheng
- 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
| | - Chen Yang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhenwei Yu
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing, China
| | - Ying Jiang
- 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
| | - 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
| |
Collapse
|
79
|
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: 2.0] [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
|
80
|
Middleton JS, Hovren HL, Kha N, Medina MJ, MacLeod KR, Concha-Marambio L, Jensen KJ. Seed amplification assay results illustrate discrepancy in Parkinson's disease clinical diagnostic accuracy and error rates. J Neurol 2023; 270:5813-5818. [PMID: 37592136 PMCID: PMC10632284 DOI: 10.1007/s00415-023-11810-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 08/19/2023]
Abstract
Parkinson's disease (PD) may be misdiagnosed due to the clinical overlap between PD and atypical parkinsonism. The utility of α-Synuclein (αSyn) Seed Amplification Assay (SAA) as a diagnostic indicator for PD has been reported in numerous studies, but never when administered as a validated clinical laboratory test. This study compares results from αSyn-SAA validation testing performed using well-characterized cohorts from two biorepositories to better understand the accuracy of PD clinical diagnosis. Blinded cerebrospinal fluid (CSF) specimens from a repository that included cohorts of subjects clinically diagnosed as PD or healthy controls, both with confirmatory dopamine transporter single-photon emission computed tomography (DAT SPECT) imaging, and blinded CSF specimens from a repository that included cohorts of subjects clinically diagnosed as PD or healthy controls based on clinical diagnosis alone, were tested as part of the validation studies for the diagnostic αSyn-SAA test (SYNTap® Biomarker Test). Measured αSyn-SAA test accuracy was 83.9% using clinical diagnosis as comparator, and 93.6% using clinical diagnosis with confirmatory DAT- SPECT imaging as comparator. The statistically significant discordance between accuracy determinations using specimens classified using different diagnostic inclusion criteria indicates that there is some symbiosis between dopamine-weighted imaging and αSyn-SAA results, both of which are associated with higher accuracy compared with the clinical diagnosis alone.
Collapse
Affiliation(s)
| | | | - Nelson Kha
- Clinical Laboratory, Amprion Inc, San Diego, CA, USA
| | | | | | - Luis Concha-Marambio
- Research Laboratory, Amprion Inc, 10355 Science Center Drive, San Diego, CA, 92121, USA
| | | |
Collapse
|
81
|
Fernandes Gomes B, Farris CM, Ma Y, Concha-Marambio L, Lebovitz R, Nellgård B, Dalla K, Constantinescu J, Constantinescu R, Gobom J, Andreasson U, Zetterberg H, Blennow K. α-Synuclein seed amplification assay as a diagnostic tool for parkinsonian disorders. Parkinsonism Relat Disord 2023; 117:105807. [PMID: 37591709 DOI: 10.1016/j.parkreldis.2023.105807] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Synucleinopathies such as Parkinson's disease (PD) and multiple system atrophy (MSA) can be challenging to diagnose due to the symptom overlap with, for example, atypical parkinsonisms like progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). Seed amplification assays (SAA), developed for the detection of α-synuclein (αSyn) aggregates in CSF, have been successful when used as a biomarker evaluation for synucleinopathies. In this study, we investigated the potential of this assay to not only detect αSyn seeds in CSF, but also discriminate between movement disorders. METHODS The αSyn-SAA was tested in a Scandinavian cohort composed of 129 CSF samples from patients with PD (n = 55), MSA (n = 27), CBD (n = 7), and PSP (n = 16), as well as healthy controls (HC, n = 24). RESULTS The αSyn seed amplification assay (αSyn-SAA) was able to correctly identify all PD samples as positive (sensitivity of 100%) while also discriminating the PD group from HC (70.8% specificity, p < 0.0001) and tauopathies [CBD (71% specificity) and PSP (75% specificity), p < 0.0001)]. The αSyn-SAA was also able to identify almost all MSA samples as positive for αSyn aggregation (sensitivity of 92.6%). In general, this assay is able to discriminate between the synucleinopathies and tauopathies analyzed herein (p < 0.0001) despite the overlapping symptoms in these diseases. CONCLUSION These findings suggest the αSyn-SAA is a useful diagnostic tool for differentiating between different parkinsonian disorders, although further optimization may be needed.
Collapse
Affiliation(s)
- Bárbara Fernandes Gomes
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | | | - Yihua Ma
- R&D Unit, Amprion Inc., San Diego, CA, 92121, USA
| | | | | | - Bengt Nellgård
- Department of Anesthesiology and Intensive Care, Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Keti Dalla
- Department of Anesthesiology and Intensive Care, Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | | | - Radu Constantinescu
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and 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 and 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; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
82
|
Frey B, Holzinger D, Taylor K, Ehrnhoefer DE, Striebinger A, Biesinger S, Gasparini L, O'Neill MJ, Wegner F, Barghorn S, Höglinger GU, Heym RG. Tau seed amplification assay reveals relationship between seeding and pathological forms of tau in Alzheimer's disease brain. Acta Neuropathol Commun 2023; 11:181. [PMID: 37964332 PMCID: PMC10644662 DOI: 10.1186/s40478-023-01676-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Abstract
Tau seed amplification assays (SAAs) directly measure the seeding activity of tau and would therefore be ideal biomarkers for clinical trials targeting seeding-competent tau in Alzheimer's disease (AD). However, the precise relationship between tau seeding measured by SAA and the levels of pathological forms of tau in the AD brain remains unknown. We developed a new tau SAA based on full-length 0N3R tau with sensitivity in the low fg/ml range and used it to characterize 103 brain samples from three independent cohorts. Tau seeding clearly discriminated between AD and control brain samples. Interestingly, seeding was absent in Progressive Supranuclear Palsy (PSP) putamen, suggesting that our tau SAA did not amplify 4R tau aggregates from PSP brain. The specificity of our tau SAA for AD brain was further supported by analysis of matched hippocampus and cerebellum samples. While seeding was detected in hippocampus from Braak stages I-II, no seeding was present in AD cerebellum that is devoid of tau inclusions. Analysis of 40 middle frontal gyrus samples encompassing all Braak stages showed that tau SAA seeding activity gradually increased with Braak stage. This relationship between seeding activity and the presence of tau inclusions in AD brain was further supported by robust correlations between tau SAA results and the levels of phosphorylated tau212/214, phosphorylated tau181, aggregated tau, and sarkosyl-insoluble tau. Strikingly, we detected tau seeding in the middle frontal gyrus already at Braak stage II-III, suggesting that tau SAA can detect tau pathology earlier than conventional immunohistochemical staining. In conclusion, our data suggest a quantitative relationship between tau seeding activity and pathological forms of tau in the human brain and provides an important basis for further development of tau SAA for accessible human samples.
Collapse
Affiliation(s)
- Bryan Frey
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany.
- Department of Neurology, Hannover Medical School, Hanover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| | - David Holzinger
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Keenan Taylor
- AbbVie Bioresearch Center, Biotherapeutics and Genetic Medicine Technologies, Worcester, MA, USA
| | - Dagmar E Ehrnhoefer
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Andreas Striebinger
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Sandra Biesinger
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Laura Gasparini
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Michael J O'Neill
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hanover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Stefan Barghorn
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Günter U Höglinger
- Department of Neurology, Hannover Medical School, Hanover, Germany
- Center for Systems Neuroscience, Hannover, Germany
- German Center for Neurodegenerative Diseases E.V. (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Roland G Heym
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany.
| |
Collapse
|
83
|
Audrain M, Egesipe AL, Tentillier N, Font L, Ratnam M, Mottier L, Clavel M, Le Roux-Bourdieu M, Fenyi A, Ollier R, Chevalier E, Guilhot F, Fuchs A, Piorkowska K, Carlyle B, Arnold SE, Berry JD, Luthi-Carter R, Adolfsson O, Pfeifer A, Kosco-Vilbois M, Seredenina T, Afroz T. Targeting amyotrophic lateral sclerosis by neutralizing seeding-competent TDP-43 in CSF. Brain Commun 2023; 5:fcad306. [PMID: 38025276 PMCID: PMC10644982 DOI: 10.1093/braincomms/fcad306] [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: 06/12/2023] [Revised: 09/08/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
In amyotrophic lateral sclerosis, a disease driven by abnormal transactive response DNA-binding protein of 43 kDa aggregation, CSF may contain pathological species of transactive response DNA-binding protein of 43 kDa contributing to the propagation of pathology and neuronal toxicity. These species, released in part by degenerating neurons, would act as a template for the aggregation of physiological protein contributing to the spread of pathology in the brain and spinal cord. In this study, a robust seed amplification assay was established to assess the presence of seeding-competent transactive response DNA-binding protein of 43 kDa species in CSF of apparently sporadic amyotrophic lateral sclerosis patients. These samples resulted in a significant acceleration of substrate aggregation differentiating the kinetics from healthy controls. In parallel, a second assay was developed to determine the level of target engagement that would be necessary to neutralize such species in human CSF by a therapeutic monoclonal antibody targeting transactive response DNA-binding protein of 43 kDa. For this, evaluation of the pharmacokinetic/pharmacodynamic effect for the monoclonal antibody, ACI-5891.9, in vivo and in vitro confirmed that a CSF concentration of ≍1100 ng/mL would be sufficient for sustained target saturation. Using this concentration in the seed amplification assay, ACI-5891.9 was able to neutralize the transactive response DNA-binding protein of 43 kDa pathogenic seeds derived from amyotrophic lateral sclerosis patient CSF. This translational work adds to the evidence of transmission of transactive response DNA-binding protein of 43 kDa pathology via CSF that could contribute to the non-contiguous pattern of clinical manifestations observed in amyotrophic lateral sclerosis and demonstrates the ability of a therapeutic monoclonal antibody to neutralize the toxic, extracellular seeding-competent transactive response DNA-binding protein of 43 kDa species in the CSF of apparently sporadic amyotrophic lateral sclerosis patients.
Collapse
Affiliation(s)
| | | | | | - Laure Font
- Research, AC Immune SA, 1015 Lausanne, Switzerland
| | | | | | | | | | - Alexis Fenyi
- Research, AC Immune SA, 1015 Lausanne, Switzerland
| | | | | | | | - Aline Fuchs
- Research, AC Immune SA, 1015 Lausanne, Switzerland
| | | | - Becky Carlyle
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Steven E Arnold
- Department of Neurology and the Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | - Tariq Afroz
- Research, AC Immune SA, 1015 Lausanne, Switzerland
| |
Collapse
|
84
|
Tropea TF, Kannarkat GT, Shaw LM. Early and Accurate Diagnosis of Parkinson Disease May Be Rooted in Seed Amplification Assays. Clin Chem 2023; 69:1209-1211. [PMID: 37688494 PMCID: PMC10857755 DOI: 10.1093/clinchem/hvad111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 09/11/2023]
Affiliation(s)
- Thomas F. Tropea
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - George T. Kannarkat
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Parkinson’s Disease Research, Education and Clinical Center, Veterans Affairs Hospital, Philadelphia, PA, United States
| | - Leslie M. Shaw
- Department of Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
85
|
Beach TG, Adler CH, Shill HA, Serrano GE, Zhang N. α-synuclein seed amplification in Parkinson's disease. Lancet Neurol 2023; 22:985. [PMID: 37863604 DOI: 10.1016/s1474-4422(23)00373-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/22/2023] [Indexed: 10/22/2023]
Affiliation(s)
- Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, USA.
| | | | - Holly A Shill
- Barrow Neurological Institute, Phoenix 85351, AZ, USA
| | | | - Nan Zhang
- Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
86
|
Bräuer S, Rossi M, Sajapin J, Henle T, Gasser T, Parchi P, Brockmann K, Falkenburger BH. Kinetic parameters of alpha-synuclein seed amplification assay correlate with cognitive impairment in patients with Lewy body disorders. Acta Neuropathol Commun 2023; 11:162. [PMID: 37814347 PMCID: PMC10563218 DOI: 10.1186/s40478-023-01653-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/17/2023] [Indexed: 10/11/2023] Open
Abstract
The alpha-synuclein (aSyn) seed amplification assay (SAA) can identify aSyn aggregates as indicator for Lewy body pathology in biomaterials of living patients and help in diagnosing Parkinson´s disease and dementia syndromes. Our objective was to confirm that qualitative results of aSyn SAA are reproducible across laboratories and to determine whether quantitative findings correlate with patient clinical characteristics. Therefore cerebrospinal fluid samples were re-analysed by aSyn SAA in a second laboratory with four technical replicates for each sample. Kinetic parameters derived from each aggregation curve were summarized and correlated with patient characteristics. We found that qualitative findings were identical between the two laboratories for 54 of 55 patient samples. The number of positive replicates for each sample also showed good agreement between laboratories. Moreover, specific kinetic parameters of the SAA showed a strong correlation with clinical parameters, notably with cognitive performance evaluated by the Montreal Cognitive Assessment. We concluded that SAA findings are highly reproducible across laboratories following the same protocol. SAA reports not only the presence of Lewy pathology but is also associated with clinical characteristics. Thus, aSyn SAA can potentially be used for patient stratification and determining the target engagement of aSyn targeting treatments.
Collapse
Affiliation(s)
- Stefan Bräuer
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Johann Sajapin
- Department of Food Chemistry, TU Dresden, Dresden, Germany
| | - Thomas Henle
- Department of Food Chemistry, TU Dresden, Dresden, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Kathrin Brockmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Björn H Falkenburger
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.
| |
Collapse
|
87
|
Mantyh WG, Block AD, Castro MR, Hansen A, Matheson MJ, Strong C, Hill A, Cayci Z, Henderson JN. Characteristics of Recurrent Visions of the Nonphysical World Among Cognitively Unimpaired Elders of the Ojibwe Tribal Nation. JAMA Netw Open 2023; 6:e2338221. [PMID: 37851441 PMCID: PMC10585403 DOI: 10.1001/jamanetworkopen.2023.38221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/28/2023] [Indexed: 10/19/2023] Open
Abstract
Importance Visual hallucinations are a core feature of dementia with Lewy bodies and primary psychiatric disease, yet identification of a hallucination vs normal spiritual experience depends on cultural context. Almost no information exists in the medical literature regarding normal spiritual experiences in American Indian participants in the context of a neurocognitive evaluation. Objective To assess the characteristics of a normal spiritual experience in an Ojibwe Tribal Nation. Design, Setting, and Participants This prospective, cross-sectional study was conducted between August 1, 2021, and August 31, 2022, among an Ojibwe Tribal Nation in northern Minnesota. Participants were evaluated at their tribal nation clinic. Cognitively unimpaired tribal Elders who were enrolled members of the tribal nation and aged 55 years or older were invited to participate via fliers, radio advertisements, and health fair presentations. Thirty-seven tribal Elders volunteered. Main Outcomes and Measures Each participant was asked whether they experienced hallucinations or visions of people, animals, or objects that are not part of the physical world. This was an a priori formulated question and part of a comprehensive neurocognitive evaluation consisting of history and physical examination (including cognitive screening with a subspecialty-trained behavioral neurologist); blood tests for metabolic, nutritional, and thyroid conditions; and noncontrast magnetic resonance imaging brain scan. Four patients were excluded from the present analysis due to having mild cognitive impairment or dementia. Results Thirty-three cognitively unimpaired tribal Elders (mean [SD] age, 66.0 [7.5] years; 22 women [67%]) were included. Sixteen (48%) answered affirmatively, reporting recurrent visions of the nonphysical world. Generally, these visions were well formed, benevolent in nature, and transient; started in preadolescence; involved spirits or ancestors; and were congruent with cultural and spiritual beliefs of the Ojibwe people. No patients had accompanying dream enactment behavior, dysautonomia, parkinsonism, sleep transition-related hallucinations, or moderate to severe depression to suggest a prodrome of an α-synucleinopathy, hypnopompic or hypnagogic hallucinations, or psychosis. Conclusions and Relevance Although based on only 1 Ojibwe Tribal Nation, this study suggests that formed visions of the nonphysical world are common among cognitively healthy Ojibwe individuals and can represent normal spiritual experiences. Clinicians would benefit from careful consideration of cultural or spiritual context to avoid misdiagnosis of neuropsychiatric disease.
Collapse
Affiliation(s)
| | - Adam D. Block
- Department of Neurology, University of Minnesota, Minneapolis
| | - Madelyn R. Castro
- Department of Neurology, University of Minnesota, Minneapolis
- Department of Psychological Sciences, Rice University, Houston, Texas
| | - Adam Hansen
- Department of Neurology, University of Minnesota, Minneapolis
| | | | - Corey Strong
- Bois Forte Health Services, Bois Forte Band of Chippewa, Nett Lake, Minnesota
| | - Annamarie Hill
- Memory Keepers Medical Discovery Team, University of Minnesota Duluth, Duluth
- Community Outreach, Strategy, External Relations, University of Minnesota Duluth, Duluth
| | - Zuzan Cayci
- Department of Radiology, University of Minnesota, Minneapolis
| | - J. Neil Henderson
- Memory Keepers Medical Discovery Team, University of Minnesota Duluth, Duluth
- Department of Family Medicine and Biobehavioral Health, University of Minnesota Duluth, Duluth
| |
Collapse
|
88
|
Talyansky S, Le Guen Y, Kasireddy N, Belloy ME, Greicius MD. APOE-ε4 and BIN1 increase risk of Alzheimer's disease pathology but not specifically of Lewy body pathology. Acta Neuropathol Commun 2023; 11:149. [PMID: 37700353 PMCID: PMC10496176 DOI: 10.1186/s40478-023-01626-6] [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: 04/25/2023] [Accepted: 07/22/2023] [Indexed: 09/14/2023] Open
Abstract
Lewy body (LB) pathology commonly occurs in individuals with Alzheimer's disease (AD) pathology. However, it remains unclear which genetic risk factors underlie AD pathology, LB pathology, or AD-LB co-pathology. Notably, whether APOE-ε4 affects risk of LB pathology independently from AD pathology is controversial. We adapted criteria from the literature to classify 4,985 subjects from the National Alzheimer's Coordinating Center (NACC) and the Rush University Medical Center as AD-LB co-pathology (AD+LB+), sole AD pathology (AD+LB-), sole LB pathology (AD-LB+), or no pathology (AD-LB-). We performed a meta-analysis of a genome-wide association study (GWAS) per subpopulation (NACC/Rush) for each disease phenotype compared to the control group (AD-LB-), and compared the AD+LB+ to AD+LB- groups. APOE-ε4 was significantly associated with risk of AD+LB- and AD+LB+ compared to AD-LB-. However, APOE-ε4 was not associated with risk of AD-LB+ compared to AD-LB- or risk of AD+LB+ compared to AD+LB-. Associations at the BIN1 locus exhibited qualitatively similar results. These results suggest that APOE-ε4 is a risk factor for AD pathology, but not for LB pathology when decoupled from AD pathology. The same holds for BIN1 risk variants. These findings, in the largest AD-LB neuropathology GWAS to date, distinguish the genetic risk factors for sole and dual AD-LB pathology phenotypes. Our GWAS meta-analysis summary statistics, derived from phenotypes based on postmortem pathologic evaluation, may provide more accurate disease-specific polygenic risk scores compared to GWAS based on clinical diagnoses, which are likely confounded by undetected dual pathology and clinical misdiagnoses of dementia type.
Collapse
Affiliation(s)
- Seth Talyansky
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA.
- Institut du Cerveau, Paris Brain Institute - ICM, Paris, France.
| | - Nandita Kasireddy
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, 290 Jane Stanford Way, E265, Stanford, CA, 94305-5090, USA
| |
Collapse
|
89
|
Meyer N, Bentin J, Janot JM, Abrao-Nemeir I, Charles-Achille S, Pratlong M, Aquilina A, Trinquet E, Perrier V, Picaud F, Torrent J, Balme S. Ultrasensitive Detection of Aβ42 Seeds in Cerebrospinal Fluid with a Nanopipette-Based Real-Time Fast Amyloid Seeding and Translocation Assay. Anal Chem 2023; 95:12623-12630. [PMID: 37587130 DOI: 10.1021/acs.analchem.3c00017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
In this work, early-stage Aβ42 aggregates were detected using a real-time fast amyloid seeding and translocation (RT-FAST) assay. Specifically, Aβ42 monomers were incubated in buffer solution with and without preformed Aβ42 seeds in a quartz nanopipette coated with L-DOPA. Then, formed Aβ42 aggregates were analyzed on flyby resistive pulse sensing at various incubation time points. Aβ42 aggregates were detected only in the sample with Aβ42 seeds after 180 min of incubation, giving an on/off readout of the presence of preformed seeds. Moreover, this RT-FAST assay could detect preformed seeds spiked in 4% cerebrospinal fluid/buffer solution. However, in this condition, the time to detect the first aggregates was increased. Analysis of Cy3-labeled Aβ42 monomer adsorption on a quartz substrate after L-DOPA coating by confocal fluorescence spectroscopy and molecular dynamics simulation showed the huge influence of Aβ42 adsorption on the aggregation process.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Jeremy Bentin
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne-Franche-Comté (UFR Sciences et Techniques), Centre Hospitalier Universitaire de Besançon, 16 route de Gray, 25030 Besançon, France
| | - Jean-Marc Janot
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Imad Abrao-Nemeir
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Saly Charles-Achille
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Maud Pratlong
- PerkinElmer, Parc Marcel Boiteux, 30200 Codolet, France
| | | | - Eric Trinquet
- PerkinElmer, Parc Marcel Boiteux, 30200 Codolet, France
| | - Veronique Perrier
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Fabien Picaud
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne-Franche-Comté (UFR Sciences et Techniques), Centre Hospitalier Universitaire de Besançon, 16 route de Gray, 25030 Besançon, France
| | - Joan Torrent
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Sebastien Balme
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| |
Collapse
|
90
|
Massey RS, McConnell EM, Chan D, Holahan MR, DeRosa MC, Prakash R. Non-invasive Monitoring of α-Synuclein in Saliva for Parkinson's Disease Using Organic Electrolyte-Gated FET Aptasensor. ACS Sens 2023; 8:3116-3126. [PMID: 37506391 DOI: 10.1021/acssensors.3c00757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Parkinson's disease (PD) currently affects more than 1 million people in the US alone, with nearly 8.5 million suffering from the disease worldwide, as per the World Health Organization. However, there remains no fast, pain-free, and effective method of screening for the disease in the ageing population, which also happens to be the most susceptible to this neurodegenerative disease. αSynuclein (αSyn) is a promising PD biomarker, demonstrating clear delineations between levels of the αSyn monomer and the extent of αSyn aggregation in the saliva of PD patients and healthy controls. In this work, we have demonstrated a laboratory prototype of a soft fluidics integrated organic electrolyte-gated field-effect transistor (OEGFET) aptasensor platform capable of quantifying levels of αSyn aggregation in saliva. The aptasensor relies on a recently reported synthetic aptamer which selectively binds to αSyn monomer as the bio-recognition molecule within the integrated fluidic channel of the biosensor. The produced saliva sensor is label-free, fast, and reusable, demonstrating good selectivity only to the target molecule in its monomer form. The novelty of these devices is the fully isolated organic semiconductor, which extends the shelf life, and the novel fully integrated soft microfluidic channels, which simplify saliva loading and testing. The OEGFET aptasensor has a limit of detection of 10 fg/L for the αSyn monomer in spiked saliva supernatant solutions, with a linear range of 100 fg/L to 10 μg/L. The linear range covers the physiological range of the αSyn monomer in the saliva of PD patients. Our biosensors demonstrate a desirably low limit of detection, an extended linear range, and fully integrated microchannels for saliva sample handling, making them a promising platform for non-invasive point-of-care testing of PD.
Collapse
Affiliation(s)
- Roslyn S Massey
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S5B6, Canada
| | - Erin M McConnell
- Department of Chemistry and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Dennis Chan
- Dept of Neuroscience, Health Sciences Building, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Matthew R Holahan
- Dept of Neuroscience, Health Sciences Building, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Maria C DeRosa
- Department of Chemistry and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1T2S2, Canada
| | - Ravi Prakash
- Department of Electronics Engineering, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
91
|
Kell DB, Pretorius E. Are fibrinaloid microclots a cause of autoimmunity in Long Covid and other post-infection diseases? Biochem J 2023; 480:1217-1240. [PMID: 37584410 DOI: 10.1042/bcj20230241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
It is now well established that the blood-clotting protein fibrinogen can polymerise into an anomalous form of fibrin that is amyloid in character; the resultant clots and microclots entrap many other molecules, stain with fluorogenic amyloid stains, are rather resistant to fibrinolysis, can block up microcapillaries, are implicated in a variety of diseases including Long COVID, and have been referred to as fibrinaloids. A necessary corollary of this anomalous polymerisation is the generation of novel epitopes in proteins that would normally be seen as 'self', and otherwise immunologically silent. The precise conformation of the resulting fibrinaloid clots (that, as with prions and classical amyloid proteins, can adopt multiple, stable conformations) must depend on the existing small molecules and metal ions that the fibrinogen may (and is some cases is known to) have bound before polymerisation. Any such novel epitopes, however, are likely to lead to the generation of autoantibodies. A convergent phenomenology, including distinct conformations and seeding of the anomalous form for initiation and propagation, is emerging to link knowledge in prions, prionoids, amyloids and now fibrinaloids. We here summarise the evidence for the above reasoning, which has substantial implications for our understanding of the genesis of autoimmunity (and the possible prevention thereof) based on the primary process of fibrinaloid formation.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
92
|
Giladi N, Alcalay RN, Cutter G, Gasser T, Gurevich T, Höglinger GU, Marek K, Pacchetti C, Schapira AHV, Scherzer CR, Simuni T, Minini P, Sardi SP, Peterschmitt MJ. Safety and efficacy of venglustat in GBA1-associated Parkinson's disease: an international, multicentre, double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Neurol 2023; 22:661-671. [PMID: 37479372 DOI: 10.1016/s1474-4422(23)00205-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/27/2023] [Accepted: 05/23/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Variants in the GBA1 gene, which encodes lysosomal acid glucocerebrosidase, are among the most common genetic risk factors for Parkinson's disease and are associated with faster disease progression. The mechanisms involved are unresolved but might include accumulation of glucosylceramide. Venglustat is a brain-penetrant glucosylceramide synthase inhibitor that, in previous studies, reduced amounts of the glycosphingolipid. We aimed to assess the safety, efficacy, and target engagement of venglustat in people with early-stage Parkinson's disease carrying pathogenic GBA1 variants. METHODS MOVES-PD part 2 was a randomised, double-blinded, placebo-controlled phase 2 study done at 52 centres (academic sites, specialty clinics, and general neurology centres) in 16 countries. Eligible adults aged 18-80 years with Parkinson's disease (Hoehn and Yahr stage ≤2) and one or more GBA1 variants were randomly assigned using an interactive voice-response system (1:1) to 52 weeks of treatment with oral venglustat (15 mg/day) or matching placebo. Investigators, site personnel, participants, and their caregivers were masked to treatment allocation. The primary outcome measure was the change from baseline to 52 weeks in the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) parts II and III combined score (a higher score indicates greater impairment), and it was analysed in a modified intention-to-treat population (ie, all randomly assigned participants with a baseline and at least one post-baseline measurement during the treatment period). This study was registered with ClinicalTrials.gov (NCT02906020) and is closed to recruitment. FINDINGS Between Dec 15, 2016, and May 27, 2021, 221 participants were randomly assigned to venglustat (n=110) or placebo (n=111). The least squares mean change in MDS-UPDRS parts II and III combined score was 7·29 (SE 1·36) for venglustat (n=96) and 4·71 (SE 1·27) for placebo (n=105); the absolute difference between groups was 2·58 (95% CI -1·10 to 6·27; p=0·17). The most common treatment-emergent adverse events (TEAEs) were constipation and nausea (both were reported by 23 [21%] of 110 participants in the venglustat group and eight [7%] of 111 participants in the placebo group). Serious TEAEs were reported for 12 (11%) participants in each group. There was one death in the venglustat group owing to an unrelated cardiopulmonary arrest and there were no deaths in the placebo group. INTERPRETATION In people with GBA1-associated Parkinson's disease in our study, venglustat had a satisfactory safety profile but showed no beneficial treatment effect compared with placebo. These findings indicate that glucosylceramide synthase inhibition with venglustat might not be a viable therapeutic approach for GBA1-associated Parkinson's disease. FUNDING Sanofi.
Collapse
Affiliation(s)
- Nir Giladi
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Centre, Sackler School of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Roy N Alcalay
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Centre, Sackler School of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel; Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY, USA
| | - Gary Cutter
- University of Alabama at Birmingham, School of Public Health, Birmingham, AL, USA
| | - Thomas Gasser
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Tanya Gurevich
- Movement Disorders Unit, Neurological Institute, Tel Aviv Sourasky Medical Centre, Sackler School of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Günter U Höglinger
- Department of Neurology, Ludwig Maximilian University, Munich, Germany; German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Claudio Pacchetti
- Parkinson's Disease and Movement Disorders Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Clemens R Scherzer
- Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Tanya Simuni
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | |
Collapse
|
93
|
Vissers MFJM, Troyer MD, Thijssen E, Pereira DR, Heuberger |JAAC, Groeneveld GJ, Huntwork‐Rodriguez S. A leucine-rich repeat kinase 2 (LRRK2) pathway biomarker characterization study in patients with Parkinson's disease with and without LRRK2 mutations and healthy controls. Clin Transl Sci 2023; 16:1408-1420. [PMID: 37177855 PMCID: PMC10432885 DOI: 10.1111/cts.13541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/15/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Increased leucine-rich repeat kinase 2 (LRRK2) kinase activity is an established risk factor for Parkinson's disease (PD), and several LRRK2 kinase inhibitors are in clinical development as potential novel disease-modifying therapeutics. This biomarker characterization study explored within- and between-subject variability of multiple LRRK2 pathway biomarkers (total LRRK2 [tLRRK2], phosphorylation of the serine 935 (Ser935) residue on LRRK2 [pS935], phosphorylation of Rab10 [pRab10], and total Rab10 [tRab10]) in different biological sources (whole blood, peripheral blood mononuclear cells [PBMCs], neutrophils) as candidate human target engagement and pharmacodynamic biomarkers for implementation in phase I/II pharmacological studies of LRRK2 inhibitors. PD patients with a LRRK2 mutation (n = 6), idiopathic PD patients (n = 6), and healthy matched control subjects (n = 10) were recruited for repeated blood and cerebrospinal fluid (CSF) sampling split over 2 days. Within-subject variability (geometric coefficient of variation [CV], %) of these biomarkers was lowest in whole blood and neutrophils (range: 12.64%-51.32%) and considerably higher in PBMCs (range: 34.81%-273.88%). Between-subject variability displayed a similar pattern, with relatively lower variability in neutrophils (range: 61.30%-66.26%) and whole blood (range: 44.94%-123.11%), and considerably higher variability in PBMCs (range: 189.60%-415.19%). Group-level differences were observed with elevated mean pRab10 levels in neutrophils and a reduced mean pS935/tLRRK2 ratio in PBMCs in PD LRRK2-mutation carriers compared to healthy controls. These findings suggest that the evaluated biomarkers and assays could be used to verify pharmacological mechanisms of action and help explore the dose-response of LRRK2 inhibitors in early-phase clinical studies. In addition, comparable α-synuclein aggregation in CSF was observed in LRRK2-mutation carriers compared to idiopathic PD patients.
Collapse
Affiliation(s)
- Maurits F. J. M. Vissers
- Centre for Human Drug ResearchLeidenThe Netherlands
- Leiden University Medical CenterLeidenThe Netherlands
| | | | - Eva Thijssen
- Centre for Human Drug ResearchLeidenThe Netherlands
- Leiden University Medical CenterLeidenThe Netherlands
| | | | | | - Geert Jan Groeneveld
- Centre for Human Drug ResearchLeidenThe Netherlands
- Leiden University Medical CenterLeidenThe Netherlands
| | | |
Collapse
|
94
|
Palmqvist S, Rossi M, Hall S, Quadalti C, Mattsson-Carlgren N, Dellavalle S, Tideman P, Pereira JB, Nilsson MH, Mammana A, Janelidze S, Baiardi S, Stomrud E, Parchi P, Hansson O. Cognitive effects of Lewy body pathology in clinically unimpaired individuals. Nat Med 2023; 29:1971-1978. [PMID: 37464059 PMCID: PMC10427420 DOI: 10.1038/s41591-023-02450-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
α-Synuclein aggregates constitute the pathology of Lewy body (LB) disease. Little is known about the effects of LB pathology in preclinical (presymptomatic) individuals, either as isolated pathology or coexisting with Alzheimer's disease (AD) pathology (β-amyloid (Aβ) and tau). We examined the effects of LB pathology using a cerebrospinal fluid α-synuclein-seed amplification assay in 1,182 cognitively and neurologically unimpaired participants from the BioFINDER study: 8% were LB positive, 26% Aβ positive (13% of those were LB positive) and 16% tau positive. LB positivity occurred more often in the presence of Aβ positivity but not tau positivity. LB pathology had independently negative effects on cross-sectional and longitudinal global cognition and memory and on longitudinal attention/executive function. Tau had cognitive effects of a similar magnitude, but these were less pronounced for Aβ. Participants with both LB and AD (Aβ and tau) pathology exhibited faster cognitive decline than those with only LB or AD pathology. LB, but not AD, pathology was associated with reduced sense of smell. Only LB-positive participants progressed to clinical LB disease over 10 years. These results are important for individualized prognosis, recruitment and choice of outcome measures in preclinical LB disease trials, but also for the design of early AD trials because >10% of individuals with preclinical AD have coexisting LB pathology.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sofia Dellavalle
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Maria H Nilsson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
95
|
Wan L, Zhu S, Chen Z, Qiu R, Tang B, Jiang H. Multidimensional biomarkers for multiple system atrophy: an update and future directions. Transl Neurodegener 2023; 12:38. [PMID: 37501056 PMCID: PMC10375766 DOI: 10.1186/s40035-023-00370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple system atrophy (MSA) is a fatal progressive neurodegenerative disease. Biomarkers are urgently required for MSA to improve the diagnostic and prognostic accuracy in clinic and facilitate the development and monitoring of disease-modifying therapies. In recent years, significant research efforts have been made in exploring multidimensional biomarkers for MSA. However, currently few biomarkers are available in clinic. In this review, we systematically summarize the latest advances in multidimensional biomarkers for MSA, including biomarkers in fluids, tissues and gut microbiota as well as imaging biomarkers. Future directions for exploration of novel biomarkers and promotion of implementation in clinic are also discussed.
Collapse
Affiliation(s)
- Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, 410008, China.
| |
Collapse
|
96
|
Coughlin DG, Irwin DJ. Fluid and Biopsy Based Biomarkers in Parkinson's Disease. Neurotherapeutics 2023; 20:932-954. [PMID: 37138160 PMCID: PMC10457253 DOI: 10.1007/s13311-023-01379-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Several advances in fluid and tissue-based biomarkers for use in Parkinson's disease (PD) and other synucleinopathies have been made in the last several years. While work continues on species of alpha-synuclein (aSyn) and other proteins which can be measured from spinal fluid and plasma samples, immunohistochemistry and immunofluorescence from peripheral tissue biopsies and alpha-synuclein seeding amplification assays (aSyn-SAA: including real-time quaking induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA)) now offer a crucial advancement in their ability to identify aSyn species in PD patients in a categorical fashion (i.e., of aSyn + vs aSyn -); to augment clinical diagnosis however, aSyn-specific assays that have quantitative relevance to pathological burden remain an unmet need. Alzheimer's disease (AD) co-pathology is commonly found postmortem in PD, especially in those who develop dementia, and dementia with Lewy bodies (DLB). Biofluid biomarkers for tau and amyloid beta species can detect AD co-pathology in PD and DLB, which does have relevance for prognosis, but further work is needed to understand the interplay of aSyn tau, amyloid beta, and other pathological changes to generate comprehensive biomarker profiles for patients in a manner translatable to clinical trial design and individualized therapies.
Collapse
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, 9444 Medical Center Drive, ECOB 03-021, MCC 0886, La Jolla, CA, 92037, USA.
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
97
|
Vascellari S, Orrù CD, Groveman BR, Parveen S, Fenu G, Pisano G, Piga G, Serra G, Oppo V, Murgia D, Perra A, Angius F, Hughson AG, Haigh CL, Manzin A, Cossu G, Caughey B. α-Synuclein seeding activity in duodenum biopsies from Parkinson's disease patients. PLoS Pathog 2023; 19:e1011456. [PMID: 37390080 DOI: 10.1371/journal.ppat.1011456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/02/2023] Open
Abstract
Abnormal deposition of α-synuclein is a key feature and biomarker of Parkinson's disease. α-Synuclein aggregates can propagate themselves by a prion-like seeding-based mechanism within and between tissues and are hypothesized to move between the intestine and brain. α-Synuclein RT-QuIC seed amplification assays have detected Parkinson's-associated α-synuclein in multiple biospecimens including post-mortem colon samples. Here we show intra vitam detection of seeds in duodenum biopsies from 22/23 Parkinson's patients, but not in 6 healthy controls by RT-QuICR. In contrast, no tau seeding activity was detected in any of the biopsies. Our seed amplifications provide evidence that the upper intestine contains a form(s) of α-synuclein with self-propagating activity. The diagnostic sensitivity and specificity for PD in this biopsy panel were 95.7% and 100% respectively. End-point dilution analysis indicated up to 106 SD50 seeding units per mg of tissue with positivity in two contemporaneous biopsies from individual patients suggesting widespread distribution within the superior and descending parts of duodenum. Our detection of α-synuclein seeding activity in duodenum biopsies of Parkinson's disease patients suggests not only that such analyses may be useful in ante-mortem diagnosis, but also that the duodenum may be a source or a destination for pathological, self-propagating α-synuclein assemblies.
Collapse
Affiliation(s)
- Sarah Vascellari
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Christina D Orrù
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| | - Bradley R Groveman
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| | - Sabiha Parveen
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| | - Giuseppe Fenu
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Giada Pisano
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Giuseppe Piga
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Giulia Serra
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Valentina Oppo
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Daniela Murgia
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Andrew G Hughson
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| | - Cathryn L Haigh
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giovanni Cossu
- S. C. Neurology and Stroke Unit, AOBrotzu, Cagliari, Italy
| | - Byron Caughey
- Laboratory of Neurological Infections and Immunity (LNII), Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Hamilton, Montana, United States
| |
Collapse
|
98
|
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
|
99
|
Lau HHC, Martinez-Valbuena I, So RWL, Mehra S, Silver NRG, Mao A, Stuart E, Schmitt-Ulms C, Hyman BT, Ingelsson M, Kovacs GG, Watts JC. The G51D SNCA mutation generates a slowly progressive α-synuclein strain in early-onset Parkinson's disease. Acta Neuropathol Commun 2023; 11:72. [PMID: 37138318 PMCID: PMC10155462 DOI: 10.1186/s40478-023-01570-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/23/2023] [Indexed: 05/05/2023] Open
Abstract
Unique strains of α-synuclein aggregates have been postulated to underlie the spectrum of clinical and pathological presentations seen across the synucleinopathies. Whereas multiple system atrophy (MSA) is associated with a predominance of oligodendroglial α-synuclein inclusions, α-synuclein aggregates in Parkinson's disease (PD) preferentially accumulate in neurons. The G51D mutation in the SNCA gene encoding α-synuclein causes an aggressive, early-onset form of PD that exhibits clinical and neuropathological traits reminiscent of both PD and MSA. To assess the strain characteristics of G51D PD α-synuclein aggregates, we performed propagation studies in M83 transgenic mice by intracerebrally inoculating patient brain extracts. The properties of the induced α-synuclein aggregates in the brains of injected mice were examined using immunohistochemistry, a conformational stability assay, and by performing α-synuclein seed amplification assays. Unlike MSA-injected mice, which developed a progressive motor phenotype, G51D PD-inoculated animals remained free of overt neurological illness for up to 18 months post-inoculation. However, a subclinical synucleinopathy was present in G51D PD-inoculated mice, characterized by the accumulation of α-synuclein aggregates in restricted regions of the brain. The induced α-synuclein aggregates in G51D PD-injected mice exhibited distinct properties in a seed amplification assay and were much more stable than those present in mice injected with MSA extract, which mirrored the differences observed between human MSA and G51D PD brain samples. These results suggest that the G51D SNCA mutation specifies the formation of a slowly propagating α-synuclein strain that more closely resembles α-synuclein aggregates associated with PD than MSA.
Collapse
Affiliation(s)
- Heather H C Lau
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
| | - Raphaella W L So
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Surabhi Mehra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
| | - Nicholas R G Silver
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Alison Mao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Erica Stuart
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
| | - Cian Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | - Martin Ingelsson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, Rm. 4KD481, 60 Leonard Ave., Toronto, ON, M5T 0S8, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
100
|
Siderowf A, Concha-Marambio L, Lafontant DE, Farris CM, Ma Y, Urenia PA, Nguyen H, Alcalay RN, Chahine LM, Foroud T, Galasko D, Kieburtz K, Merchant K, Mollenhauer B, Poston KL, Seibyl J, Simuni T, Tanner CM, Weintraub D, Videnovic A, Choi SH, Kurth R, Caspell-Garcia C, Coffey CS, Frasier M, Oliveira LMA, Hutten SJ, Sherer T, Marek K, Soto C. Assessment of heterogeneity among participants in the Parkinson's Progression Markers Initiative cohort using α-synuclein seed amplification: a cross-sectional study. Lancet Neurol 2023; 22:407-417. [PMID: 37059509 PMCID: PMC10627170 DOI: 10.1016/s1474-4422(23)00109-6] [Citation(s) in RCA: 313] [Impact Index Per Article: 156.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Emerging evidence shows that α-synuclein seed amplification assays (SAAs) have the potential to differentiate people with Parkinson's disease from healthy controls. We used the well characterised, multicentre Parkinson's Progression Markers Initiative (PPMI) cohort to further assess the diagnostic performance of the α-synuclein SAA and to examine whether the assay identifies heterogeneity among patients and enables the early identification of at-risk groups. METHODS This cross-sectional analysis is based on assessments done at enrolment for PPMI participants (including people with sporadic Parkinson's disease from LRRK2 and GBA variants, healthy controls, prodromal individuals with either rapid eye movement sleep behaviour disorder (RBD) or hyposmia, and non-manifesting carriers of LRRK2 and GBA variants) from 33 participating academic neurology outpatient practices worldwide (in Austria, Canada, France, Germany, Greece, Israel, Italy, the Netherlands, Norway, Spain, the UK, and the USA). α-synuclein SAA analysis of CSF was performed using previously described methods. We assessed the sensitivity and specificity of the α-synuclein SAA in participants with Parkinson's disease and healthy controls, including subgroups based on genetic and clinical features. We established the frequency of positive α-synuclein SAA results in prodromal participants (RBD and hyposmia) and non-manifesting carriers of genetic variants associated with Parkinson's disease, and compared α-synuclein SAA to clinical measures and other biomarkers. We used odds ratio estimates with 95% CIs to measure the association between α-synuclein SAA status and categorical measures, and two-sample 95% CIs from the resampling method to assess differences in medians between α-synuclein SAA positive and negative participants for continuous measures. A linear regression model was used to control for potential confounders such as age and sex. FINDINGS This analysis included 1123 participants who were enrolled between July 7, 2010, and July 4, 2019. Of these, 545 had Parkinson's disease, 163 were healthy controls, 54 were participants with scans without evidence of dopaminergic deficit, 51 were prodromal participants, and 310 were non-manifesting carriers. Sensitivity for Parkinson's disease was 87·7% (95% CI 84·9-90·5), and specificity for healthy controls was 96·3% (93·4-99·2). The sensitivity of the α-synuclein SAA in sporadic Parkinson's disease with the typical olfactory deficit was 98·6% (96·4-99·4). The proportion of positive α-synuclein SAA was lower than this figure in subgroups including LRRK2 Parkinson's disease (67·5% [59·2-75·8]) and participants with sporadic Parkinson's disease without olfactory deficit (78·3% [69·8-86·7]). Participants with LRRK2 variant and normal olfaction had an even lower α-synuclein SAA positivity rate (34·7% [21·4-48·0]). Among prodromal and at-risk groups, 44 (86%) of 51 of participants with RBD or hyposmia had positive α-synuclein SAA (16 of 18 with hyposmia, and 28 of 33 with RBD). 25 (8%) of 310 non-manifesting carriers (14 of 159 [9%] LRRK2 and 11 of 151 [7%] GBA) were positive. INTERPRETATION This study represents the largest analysis so far of the α-synuclein SAA for the biochemical diagnosis of Parkinson's disease. Our results show that the assay classifies people with Parkinson's disease with high sensitivity and specificity, provides information about molecular heterogeneity, and detects prodromal individuals before diagnosis. These findings suggest a crucial role for the α-synuclein SAA in therapeutic development, both to identify pathologically defined subgroups of people with Parkinson's disease and to establish biomarker-defined at-risk cohorts. FUNDING PPMI is funded by the Michael J Fox Foundation for Parkinson's Research and funding partners, including: Abbvie, AcureX, Aligning Science Across Parkinson's, Amathus Therapeutics, Avid Radiopharmaceuticals, Bial Biotech, Biohaven, Biogen, BioLegend, Bristol-Myers Squibb, Calico Labs, Celgene, Cerevel, Coave, DaCapo Brainscience, 4D Pharma, Denali, Edmond J Safra Foundation, Eli Lilly, GE Healthcare, Genentech, GlaxoSmithKline, Golub Capital, Insitro, Janssen Neuroscience, Lundbeck, Merck, Meso Scale Discovery, Neurocrine Biosciences, Prevail Therapeutics, Roche, Sanofi Genzyme, Servier, Takeda, Teva, UCB, VanquaBio, Verily, Voyager Therapeutics, and Yumanity.
Collapse
Affiliation(s)
- Andrew Siderowf
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | | | - David-Erick Lafontant
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Carly M Farris
- Research and Development Unit, Amprion, San Diego, CA, USA
| | - Yihua Ma
- Research and Development Unit, Amprion, San Diego, CA, USA
| | - Paula A Urenia
- Research and Development Unit, Amprion, San Diego, CA, USA
| | - Hieu Nguyen
- Research and Development Unit, Amprion, San Diego, CA, USA
| | - Roy N Alcalay
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Douglas Galasko
- Department of Neurology, University of California, San Diego, CA, USA
| | - Karl Kieburtz
- University of Rochester Medical Center, University of Rochester, Rochester, NY, USA
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany; Paracelsus-Elena Klinik, Kassel, and German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Caroline M Tanner
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA; Parkinson's Disease Research, Education and Clinical Center, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Parkinson's Disease Research, Education and Clinical Center, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Aleksandar Videnovic
- Department of Neurology, Neurological Clinical Research Institute, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Seung Ho Choi
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Ryan Kurth
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Chelsea Caspell-Garcia
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Christopher S Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Mark Frasier
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Luis M A Oliveira
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Samantha J Hutten
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Todd Sherer
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Claudio Soto
- Research and Development Unit, Amprion, San Diego, CA, USA; Department of Neurology, University of Texas McGovern Medical School at Houston, TX, USA
| |
Collapse
|