1
|
Brooker SM, Gonzalez-Latapi P. Biomarkers in Parkinson's Disease. Neurol Clin 2025; 43:229-248. [PMID: 40185520 DOI: 10.1016/j.ncl.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Parkinson's disease (PD) is a leading cause of disability worldwide, and there is a pressing need to develop therapeutics to slow or halt disease progression. The identification of reliable biomarkers of PD at all stages of disease will be a critical step toward optimizing diagnosis and therapeutic development. For PD, biomarkers could serve multiple important functions. There have been significant advances in biomarker development in PD in recent years, and in this review, the authors summarize the current state of the PD biomarker field covering major advances in fluid, tissue, and neuroimaging biomarkers.
Collapse
Affiliation(s)
- Sarah M Brooker
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paulina Gonzalez-Latapi
- The Ken and Ruth Davee Neurology Department, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
2
|
Kou W, Li S, Yan R, Zhang J, Wan Z, Feng T. Cerebrospinal fluid and blood neurofilament light chain in Parkinson's disease and atypical parkinsonian syndromes: a systematic review and Bayesian network meta-analysis. J Neurol 2025; 272:311. [PMID: 40180649 DOI: 10.1007/s00415-025-13051-x] [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: 02/08/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND AND OBJECTIVE The value of neurofilament light chain (NfL) levels as a biomarker for the diagnosis and differential diagnosis in patients with Parkinson's disease (PD) and atypical parkinsonian syndromes (APS) remains controversial. Furthermore, few studies have directly compared NfL levels among specific APS categories. This study aimed to compare cerebrospinal fluid (CSF) and blood NfL levels among PD, APS, other PD-related disorders, and controls, as well as rank NfL levels across these groups. METHODS PubMed, Embase, Web of Science, and the Cochrane Library were searched from the inception up to November 1st, 2024, to identify eligible studies reporting CSF or blood NfL concentrations in PD, PD dementia (PDD), multiple system atrophy (MSA), progressive supranuclear palsy (PSP), dementia with Lewy bodies (DLB), corticobasal syndrome (CBS), vascular parkinsonism (VP), essential tremor (ET), idiopathic rapid eye movement sleep behavior disorder (iRBD), and controls. The Bayesian approach was utilized to estimate the standardized mean difference (SMD) and the associated 95% credible intervals (CrIs) of NfL levels. The surface under the cumulative ranking curve (SUCRA) was employed to evaluate the ranking probabilities of NfL levels. Subgroup analysis and meta-regression were conducted to explore the sources of heterogeneity. RESULTS The present network meta-analysis (NMA) included 78 studies with 13,120 participants (4050 controls, 5021 PD, 191 PDD, 1173 MSA, 887 PSP, 1254 DLB, 319 CBS, 160 ET, 65 iRBD, and 0 VP). Of these, the NMA of CSF NfL included 34 studies with 6,013 participants, while the NMA of blood NfL included 49 studies with 7,787 participants. Both CSF and blood NfL levels were significantly elevated in patients with PD and APS compared to controls. Compared to PD patients, CSF NfL levels were significantly elevated in MSA (SMD 1.85; 95% CrI 1.55-2.15), CBS (1.42; 1.08-1.75), PSP (1.35; 1.06-1.64), and DLB 0.52; 0.20-0.85) patients. Similarly, blood NfL levels were significantly higher in patients with MSA (1.36; 1.02-1.71), PDD (1.19; 0.65-1.72), PSP (1.15; 0.77-1.54), CBS (0.92; 0.11-1.72), and DLB (0.63; 0.14-1.12) compared to PD. Among APS, CSF NfL levels in MSA patients were significantly higher than those in PSP, DLB, and CBS patients, while blood NfL levels in MSA patients were significantly higher only compared to DLB. In both CSF and blood NfL, MSA patients exhibited the highest probability of ranking first for NfL level elevations (CSF: SUCRA = 0.998; blood: SUCRA = 0.925). Age significantly influenced the SMD of the comparison between MSA and PD in CSF NfL (β = -0.15; p = 0.016). CONCLUSIONS CSF and blood NfL levels in PD and APS are higher than those in controls, and all APS categories show higher levels than PD, suggesting that NfL levels may serve as a potential biomarker for the differential diagnosis between PD and APS. However, caution is warranted when using NfL as a diagnostic biomarker for PD. Significant differences in NfL levels are also observed between certain APS categories. Patients with MSA exhibit the highest NfL levels among PD and related disorders.
Collapse
Affiliation(s)
- Wenyi Kou
- 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
| | - Rui Yan
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junjiao Zhang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhirong Wan
- Department of Neurology, Aerospace Center Hospital, Beijing, 100049, People's Republic of 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
|
3
|
Pedersen CC, Maple-Grødem J, Lange J. A systematic review of biofluid phosphorylated α-synuclein in Parkinson's disease. Parkinsonism Relat Disord 2025; 132:107240. [PMID: 39721932 DOI: 10.1016/j.parkreldis.2024.107240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is a progressive neurodegenerative disease, and biomarkers are needed to enhance earlier detection and monitoring. Alpha-synuclein, phosphorylated at serine 129 (pS129-α-syn), is the predominant form of α-syn found in Lewy bodies implicating an involvement in disease pathology. This review aims to systematically evaluate the evidence for pS129-α-syn detection in human biofluid samples of PD utilizing ELISA-based protein detection methods. METHODS A systematic review was conducted following the Preferred Reported Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Electronic searches were performed in PubMed, Web of Science, and Cochrane databases from inception to November 7th, 2024, to identify studies comparing pS129-α-syn in biofluids of PD patients with controls or related neurodegenerative disease. Risk of bias was assessed for each study. RESULTS Twenty-three publications met the inclusion criteria, with pS129-α-syn detected in cerebrospinal fluid, plasma, red blood cells, serum, and saliva exosomes. Overall, pS129-α-syn levels were elevated in patients with PD compared to controls, and in some studies, correlated with disease severity. There was no consistent pattern when comparing PD patients to those with related neurodegenerative diseases. Significant variability in pS129-α-syn levels and considerable overlap between groups may limit the utility as a biomarker. CONCLUSION While pS129-α-syn for PD shows some promise as a diagnostic marker for PD, its differential diagnostic utility remains limited. Further research involving larger cohorts is required. The greatest potential for pS129-α-syn may be as part of a panel with other PD-specific markers, to enhance diagnostic accuracy and prognostic value.
Collapse
Affiliation(s)
- Camilla Christina Pedersen
- Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| | - Jodi Maple-Grødem
- Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| | - Johannes Lange
- Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| |
Collapse
|
4
|
Louis ED, Sharma VD, Hajjar I, Hernandez N. Serum Neurofilament Light-Chain Levels in Essential Tremor: A Replication Study. NEURODEGENER DIS 2024; 24:141-147. [PMID: 39626650 DOI: 10.1159/000542922] [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: 10/17/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
INTRODUCTION Essential tremor (ET) is a highly prevalent neurological disease. At present, there are no clinical biomarkers. Neurofilament light (NfL) has been studied as a measure of neuronal damage in a considerable number of neurological disorders. There have been three studies of ET, and results are inconsistent. METHODS Forty ET cases were enrolled in a research study between February and November 2023 and compared to two control groups from study 1 (n = 41) and study 2 (n = 185). Total tremor score was a measure of the severity of action tremor. Blood samples were analyzed for serum NfL level on the Simoa® platform using an NF-Light™ kit as a marker of axonal injury. RESULTS Serum log NfL levels were higher in ET cases than controls in study 1 (p < 0.001) and study 2 (p < 0.001). In a multivariate linear regression model, ET cases (p = 0.03) and individuals of older age (p < 0.001) had higher serum log NfL levels than controls (combined in studies 1 and 2). There was no association in ET cases between serum log NfL level and total tremor score (Pearson's r = 0.08, p = 0.63). CONCLUSION This new study further validates the elevation in serum NfL levels in ET, now representing the third study to do so. In combination, the converging data suggest that there is an overall increase in serum NfL levels in ET. The demonstration of elevated serum levels of NfL in ET adds an additional piece of evidence that there is neuronal damage in ET.
Collapse
Affiliation(s)
- Elan D Louis
- Department of Neurology, University of Texas Southwestern, Dallas, Texas, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vibhash D Sharma
- Department of Neurology, University of Texas Southwestern, Dallas, Texas, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ihab Hajjar
- Department of Neurology, University of Texas Southwestern, Dallas, Texas, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nora Hernandez
- Department of Neurology, University of Texas Southwestern, Dallas, Texas, USA
| |
Collapse
|
5
|
Pedersen CC, Ushakova A, Alves G, Tysnes OB, Blennow K, Zetterberg H, Maple-Grødem J, Lange J. Serum neurofilament light at diagnosis: a prognostic indicator for accelerated disease progression in Parkinson's Disease. NPJ Parkinsons Dis 2024; 10:162. [PMID: 39164268 PMCID: PMC11336184 DOI: 10.1038/s41531-024-00768-1] [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: 02/26/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Neurofilament light chain (NFL) is elevated in neurodegenerative diseases, including Parkinson's disease (PD). This study aimed to investigate serum NFL in newly diagnosed PD and its association with cognitive and motor decline over 10 years. Serum NFL levels were measured in PD patients and controls from the ParkWest study at diagnosis (baseline) and after 3 and 5 years. Mixed-effects regression analyzed changes in NFL and the association with annual changes in MMSE and UPDRS-III scores over 10 years. PD patients had elevated serum NFL at all visits and a faster annual increase over 5 years compared to controls (0.09 pg/mL per year; p = 0.029). Higher baseline NFL predicted faster cognitive decline β -0.77 transformed MMSE; p = 0.010), and a 40% NFL increase predicted future motor decline (β 0.28 UPDRS-III; p = 0.004). Elevated serum NFL in early PD is linked to faster cognitive and motor impairment, suggesting its prognostic value in PD biomarker panels.
Collapse
Grants
- 287842 Norges Forskningsråd (Research Council of Norway)
- 177966 Norges Forskningsråd (Research Council of Norway)
- 287842 Norges Forskningsråd (Research Council of Norway)
- 287842 Norges Forskningsråd (Research Council of Norway)
- 29604 Western Norway Regional Health Authority | Stavanger Universitetssjukehus (Stavanger University Hospital (SUS))
- 911218 Western Norway Regional Health Authority | Stavanger Universitetssjukehus (Stavanger University Hospital (SUS))
- 29604 Helse Vest (Western Norway Regional Health Authority)
- #2017-00915 Vetenskapsrådet (Swedish Research Council)
- #2022-00732 Vetenskapsrådet (Swedish Research Council)
- #2023-00356 Vetenskapsrådet (Swedish Research Council)
- #2022-01018 Vetenskapsrådet (Swedish Research Council)
- #2019-02397 Vetenskapsrådet (Swedish Research Council)
- No 860197 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 Marie Skłodowska-Curie Actions (H2020 Excellent Science - Marie Skłodowska-Curie Actions)
- Norwegian Health Association (14846 and 16152) Rebergs legacy (no grant number)
- KB is also supported by the Swedish Alzheimer Foundation (#AF-930351, #AF-939721, #AF-968270, and #AF-994551), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236), the Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495), the Alzheimer’s Association 2022-2025 Grant (SG-23-1038904 QC), La Fondation Recherche Alzheimer (FRA), Paris, France, and the Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark.
- Cure Alzheimer's Fund (Alzheimer's Disease Research Foundation)
- Familjen Erling-Perssons Stiftelse (Erling-Persson Family Foundation)
- HZ is a Wallenberg Scholar and is also supported by grants from the European Union’s Horizon Europe research and innovation programme under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer's Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), the National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL (UKDRI-1003).
- Norwegian Health Association (16152)
Collapse
Affiliation(s)
- Camilla Christina Pedersen
- The Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Anastasia Ushakova
- Section of Biostatistics, Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Guido Alves
- The Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
- Department of Neurology, Stavanger University Hospital, Stavanger, Norway
| | - Ole-Bjørn Tysnes
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - 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
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - 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, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jodi Maple-Grødem
- The Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Johannes Lange
- The Centre for Movement Disorders, Stavanger University Hospital, Stavanger, Norway.
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| |
Collapse
|
6
|
Salinas M, Salinas VH, McGurn M, Hernandez N, Louis ED. Serum Neurofilament Light-Chain Concentrations in Essential Tremor: a Case-Control Study. CEREBELLUM (LONDON, ENGLAND) 2024; 23:951-956. [PMID: 37466893 DOI: 10.1007/s12311-023-01583-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/20/2023]
Abstract
Essential tremor (ET) is a common neurological disorder, with clinical and pathophysiological links to the cerebellum. Inquiries into the etiology, pathophysiology, and nosology of ET stand to benefit from the identification of disease biomarkers. Serum neurofilament light chain (NfL) has emerged as a novel signature of conditions in which neuronal injury reflects an outcome of the ongoing disease process. We sought to investigate the concentrations of NfL in ET patients and healthy controls. In this case-control study, our powered study population of 41 ET patients and 40 age-matched healthy controls underwent clinical assessments and measurement of serum NfL concentration using Simoa technology. Serum NfL was elevated in ET patients - mean log-transformed serum NfL concentration = 1.23 ± 0.19 (95% confidence interval [CI] = 1.17-1.29) vs. 1.08 ± 0.15 (95% CI = 1.03-1.13), p = 0.0002. This difference persisted after accounting for age, sex and Montreal Cognitive Assessment score in a multiple linear regression model (p = 0.002) and in an age-matched sample subset of 35 ET cases and 35 controls (p = 0.006). There was no association between tremor severity and serum NfL levels (p = 0.73). In this sample of ET patients and controls, serum NfL concentrations were significantly higher in ET. Studies in additional cohorts of ET cases would be of value in attempting to replicate these results and assessing diagnostic utility.
Collapse
Affiliation(s)
- Meagen Salinas
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA.
- Neurology Section, VA North Texas Health Care System, Dallas, TX, USA.
| | - Victor H Salinas
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
- Neurology Section, VA North Texas Health Care System, Dallas, TX, USA
| | - Maisie McGurn
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Nora Hernandez
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Elan D Louis
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| |
Collapse
|
7
|
Agnello L, Gambino CM, Ciaccio AM, Masucci A, Vassallo R, Tamburello M, Scazzone C, Lo Sasso B, Ciaccio M. Molecular Biomarkers of Neurodegenerative Disorders: A Practical Guide to Their Appropriate Use and Interpretation in Clinical Practice. Int J Mol Sci 2024; 25:4323. [PMID: 38673907 PMCID: PMC11049959 DOI: 10.3390/ijms25084323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Neurodegenerative disorders (NDs) represent a group of different diseases characterized by the progressive degeneration and death of the nervous system's cells. The diagnosis is challenging, especially in the early stages, due to no specific clinical signs and symptoms. In this context, laboratory medicine could support clinicians in detecting and differentiating NDs. Indeed, biomarkers could indicate the pathological mechanisms underpinning NDs. The ideal biofluid for detecting the biomarkers of NDs is cerebrospinal fluid (CSF), which has limitations, hampering its widespread use in clinical practice. However, intensive efforts are underway to introduce high-sensitivity analytical methods to detect ND biomarkers in alternative nonivasive biofluid, such as blood or saliva. This study presents an overview of the ND molecular biomarkers currently used in clinical practice. For some diseases, such as Alzheimer's disease or multiple sclerosis, biomarkers are well established and recommended by guidelines. However, for most NDs, intensive research is ongoing to identify reliable and specific biomarkers, and no consensus has yet been achieved.
Collapse
Affiliation(s)
- Luisa Agnello
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
| | - Caterina Maria Gambino
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Anna Maria Ciaccio
- Internal Medicine and Medical Specialties “G. D’Alessandro”, Department of Health Promotion, Maternal and Infant Care, University of Palermo, 90127 Palermo, Italy;
| | - Anna Masucci
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
| | - Roberta Vassallo
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
| | - Martina Tamburello
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
| | - Concetta Scazzone
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
| | - Bruna Lo Sasso
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Marcello Ciaccio
- Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.A.); (C.M.G.); (A.M.); (R.V.); (M.T.); (C.S.); (B.L.S.)
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
8
|
Ou R, Liu K, Lin J, Yang T, Xiao Y, Wei Q, Hou Y, Li C, Zhang L, Jiang Z, Zhao B, Chen X, Song W, Wu Y, Shang H. Relationship between plasma NFL and disease progression in Parkinson's disease: a prospective cohort study. J Neurol 2024; 271:1837-1843. [PMID: 38063869 DOI: 10.1007/s00415-023-12117-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 03/28/2024]
Abstract
OBJECTIVE We aimed to examine the longitudinal change of plasma neurofilament light chain (NFL) level and explore its diagnostic and prognostic implications in Parkinson's disease (PD). METHODS A total of 184 patients with early PD who completed 5-year annually repeated clinical assessments were included. Plasma NFL at baseline, 1 year, and 2 year were examined, which were quantified using the ultrasensitive Simoa technology. At baseline, blood from 86 sex- and age-matched healthy controls (HC) were obtained for comparison. RESULTS Plasma NFL in PD patients at baseline was significantly higher than those in HC (P = 0.046), and significantly increased after 2 years (P = 0.046). Receiver operating characteristic curve indicated that a plasma NFL cut-off value of 10.79 pg/mL resulted in 39.7% sensitivity and 84.0% specificity, with an area under the curve of 0.635, to distinguish PD from HC (P < 0.001). Linear mixed-effect models indicated that baseline plasma NFL (> 9.24 pg/mL) correlated with a greater increase in the Unified Parkinson's Disease Rating Scale III (estimate = 0.651, P = 0.001) and Hoehn & Yahr stage (estimate = 0.072, P < 0.001), and also correlated with a greater decrease in the Montreal Cognitive Assessment (estimate = - 0.387, P < 0.001) during follow-up visits. CONCLUSIONS Plasma NFL exhibits a tendency to increase with disease progression, and elevated baseline plasma NFL can serve as a predictor for accelerated motor deterioration and cognitive decline in PD. However, plasma NFL does not have high accuracy to distinguish individuals with early-stage PD from HC.
Collapse
Affiliation(s)
- Ruwei Ou
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Kuncheng Liu
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Junyu Lin
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Tianmi Yang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yi Xiao
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Qianqian Wei
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yanbing Hou
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Chunyu Li
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Lingyu Zhang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Bi Zhao
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Xueping Chen
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Wei Song
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Ying Wu
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Huifang Shang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.37, Guoxue Lane, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
9
|
Kong W, Li X, Guo X, Sun Y, Chai W, Chang Y, Huang Q, Wang P, Wang X. Ultrasound-Assisted CRISPRi-Exosome for Epigenetic Modification of α-Synuclein Gene in a Mouse Model of Parkinson's Disease. ACS NANO 2024; 18:7837-7851. [PMID: 38437635 DOI: 10.1021/acsnano.3c05864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Currently, there is a lack of effective treatment for Parkinson's disease (PD). In PD patients, aberrant methylation of SNCA (α-synuclein gene) has been reported and may be a potential therapeutic target. In this study, we established an epigenetic regulation platform based on an exosomal CRISPR intervention system. With the assist of focused ultrasound (FUS) opening the blood-brain barrier, engineered exosomes carrying RVG (rabies viral glycoprotein) targeting peptide, sgRNA (single guide RNA), and dCas9-DNMT3A (named RVG-CRISPRi-Exo) were efficiently delivered into the brain lesions and induced specific methylation of SNCA. In vivo, FUS combined with RVG-CRISPRi-Exo significantly improved motor performance, balance coordination, and neurosensitivity in PD mice, greatly down-regulated the elevation of α-synuclein (α-syn) caused by modeling, rescued cell apoptosis, and alleviated the progression of PD in mice. [18F]-FP-DTBZ imaging suggested that the synaptic function of the nigrostriatal pathway could be restored, which was conducive to the control of motor behavior in PD mice. Pyrosequencing results showed that RVG-CRISPRi-Exo could methylate CpG at specific sites of SNCA, and this fine-tuned editing achieved good therapeutic effects in PD model mice. In vitro, RVG-CRISPRi-Exo down-regulated SNCA transcripts and α-syn expression and relieved neuronal cell damage. Collectively, our findings provide a proof-of-principle for the development of targeted brain nanodelivery based on engineered exosomes and provide insights into epigenetic regulation of brain diseases.
Collapse
Affiliation(s)
- Weirong Kong
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Xin Li
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Xiaoyu Guo
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Yue Sun
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Wenyu Chai
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Yawei Chang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Qichao Huang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Pan Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, #620 West Chang'an Road, Xi'an 710119, China
| |
Collapse
|
10
|
Brown Q, Nicholson E, Wang C, Greenlee J, Seger H, Veneziano S, Cassmann E. Temporal serum neurofilament light chain concentrations in sheep inoculated with the agent of classical scrapie. PLoS One 2024; 19:e0299038. [PMID: 38394122 PMCID: PMC10889644 DOI: 10.1371/journal.pone.0299038] [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: 09/27/2023] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Neurofilament light chain (Nf-L) has been used to detect neuroaxonal damage in the brain caused by physical injury or disease. The purpose of this study was to determine if serum Nf-L could be used as a biomarker for pre-symptomatic detection of scrapie in sheep. METHODS Four sheep with prion protein genotype AVQQ were intranasally inoculated with the classical scrapie strain x124. Blood was collected every 4 weeks until 44 weeks post-inoculation, at which point weekly collection commenced. Serum was analyzed using single molecule array (Quanterix SR-X) to evaluate Nf-L concentrations. RESULTS Scrapie was confirmed in each sheep by testing homogenized brainstem at the level of the obex with a commercially available enzyme immunoassay. Increased serum Nf-L concentrations were identified above the determined cutoff during the last tenth of the respective incubation period for each sheep. Throughout the time course study, PrPSc accumulation was not detected antemortem by immunohistochemistry in rectal tissue at any timepoint for any sheep. RT-QuIC results were inconsistently positive throughout the timepoints tested for each sheep; however, each sheep had at least one timepoint detected positive. When assessing serum Nf-L utility using receiver operator characteristic curves against different clinical parameters, such as asymptomatic and symptomatic (pruritus or neurologic signs), results showed that Nf-L was most useful at being an indicator of disease only late in disease progression when neurologic signs were present. CONCLUSION Serum Nf-L concentrations in the cohort of sheep increased as disease progressed; however, serum Nf-L did not increase during the presymptomatic window. The levels increased substantially throughout the final 10% of the animals' scrapie incubation period when other clinical signs were present. Serum Nf-L is not a reliable biomarker for pre-clinical detection of scrapie.
Collapse
Affiliation(s)
- Quazetta Brown
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Christensen, Ames, United States of America
| | - Eric Nicholson
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Justin Greenlee
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Hannah Seger
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Susan Veneziano
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| | - Eric Cassmann
- United States Department of Agriculture, Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, Iowa, United States of America
| |
Collapse
|
11
|
Soni R, Mathur K, Shah J. An update on new-age potential biomarkers for Parkinson's disease. Ageing Res Rev 2024; 94:102208. [PMID: 38296162 DOI: 10.1016/j.arr.2024.102208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that deals with dopaminergic deficiency in Substantia nigra pars compact (SNpc) region of the brain. Dopaminergic deficiency manifests into motor dysfunction. Alpha-synuclein protein aggregation is the source for inception of the pathology. Motor symptoms include rigidity, akinesia, tremor and gait dysfunction. Pre-motor symptoms are also seen in early stage of the disease; however, they are not distinguishable. Lack of early diagnosis in PD pathology poses a major challenge for development of disease modifying therapeutics. Substantial neuronal loss has already been occurred before the clinical manifestations appear and hence, it becomes impossible to halt the disease progression. Current diagnostics are majorly based on the clinical symptoms and thus fail to detect early progression of the disease. Thus, there is need for early diagnosis of PD, for detection of the disease at its inception. This will facilitate the effective use of therapies that halt the progression and will make remission possible. Many novel biomarkers are being developed that include blood-based biomarker, CSF biomarker. Other than that, there are non-invasive techniques that can detect biomarkers. We aim to discuss potential role of these new age biomarkers and their association with PD pathogenesis in this review.
Collapse
Affiliation(s)
- Ritu Soni
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Kirti Mathur
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
12
|
Pilotto A, Zanusso G, Antelmi E, Okuzumi A, Zatti C, Lupini A, Bongianni M, Padovani A, Hattori N. Biofluid Markers and Tissue Biopsies Analyses for the Prodromal and Earliest Phase of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S333-S344. [PMID: 39331105 PMCID: PMC11494635 DOI: 10.3233/jpd-240007] [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: 08/09/2024] [Indexed: 09/28/2024]
Abstract
The recent development of new methods to detect misfolded α-synuclein (αSyn) aggregates in biofluids and tissue biopsies in the earliest Parkinson's disease (PD) phases is dramatically challenging the biological definition of PD. The αSyn seed amplification methods in cerebrospinal fluid (CSF) showed high sensitivity and specificity for early diagnosis of PD and Lewy bodies disorders. Several studies in isolated REM sleep behavior disorders and other at-risk populations also demonstrated a high prevalence of CSF αSyn positivity and its potential value in predicting the phenoconversion to clinically manifested diseases. Growing evidence exists for αSyn aggregates in olfactory mucosa, skin, and other tissues in subjects with PD or at-risk subjects. DOPA decarboxylase and numerous other candidates have been additionally proposed for either diagnostic or prognostic purposes in earliest PD phases. The newly described αSyn detection in blood, through its quantification in neuronally-derived exosome vesicles, represents a technical challenge that could open a new scenario for the biological diagnosis of PD. Despite this growing evidence in the field, most of method of αSyn detection and markers still need to be validated in ongoing longitudinal studies through an accurate assessment of different prodromal disease subtypes and scenarios before being definitively implemented in clinical settings.
Collapse
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gianluigi Zanusso
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Elena Antelmi
- Neurology Unit, Parkinson Disease and Movement Disorders Division, Department of Engineering and Medicine of Innovation, University of Verona, Verona, Italy
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Cinzia Zatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Alessandro Lupini
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Matilde Bongianni
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
13
|
Novobilský R, Bartova P, Lichá K, Bar M, Stejskal D, Kusnierova P. Serum neurofilament light chain levels in patients with cognitive deficits and movement disorders: comparison of cerebrospinal and serum neurofilament light chain levels with other biomarkers. Front Hum Neurosci 2023; 17:1284416. [PMID: 38164192 PMCID: PMC10757912 DOI: 10.3389/fnhum.2023.1284416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Background Serum neurofilament light chain (S NfL) is a non-specific marker of neuronal damage, including Alzheimer's disease (AD). We aimed to verify the reference interval (RI) of serum NfL using a highly sensitive ELISA, and to estimate the optimal cut-off value for neuronal damage. Our second objective was to compare NfL in cerebrospinal fluid (CSF) and serum (S) with the routine neurodegeneration biomarkers used in AD, and to assess their concentrations relative to the degree of cognitive deficit. Methods Samples from 124 healthy volunteers were used to estimate the S NfL RI. For the comparison study, we used CSF and S samples from 112 patients with cognitive disorders. Cognitive functions were assessed using the mini-mental state examination. ELISA assays were used to determine the CSF and S NfL levels, CSF β-amyloid peptide42 (Aβ42), CSF β-amyloid peptide40 (Aβ40), CSF total tau protein (tTau), CSF phosphorylated tau protein (pTau), and CSF alpha-synuclein (αS). Results The estimated RI of S NfL were 2.25-9.19 ng.L-1. The cut-off value of S NfL for assessing the degree of neuronal impairment was 10.5 ng.L-1. We found a moderate statistically significant correlation between S NfL and CSF Aβ42 in the group with movement disorders, without dementia (rs = 0.631; p = 0.016); between S NfL and CSF Aβ40 in the group with movement disorder plus dementia (rs = -0.750; p = 0.052); between S NfL and CSF tTau in the control group (rs = 0.689; p = 0.009); and between S NfL and CSF pTau in the control group (rs = 0.749; p = 0.003). The non-parametric Kruskal-Wallis test revealed statistically significant differences between S NfL, CSF NfL, CSF Aβ42, CSF tTau, and CSF pTau and diagnosis within groups. The highest kappa coefficients were found between the concentrations of S NfL and CSF NfL (κ = 0.480) and between CSF NfL and CSF tTau (κ = 0.351). Conclusion Our results suggested that NfL and tTau in CSF of patients with cognitive decline could be replaced by the less-invasive determination of S NfL using a highly sensitive ELISA method. S NfL reflected the severity of cognitive deficits assessed by mini-mental state examination (MMSE). However, S NfL is not specific to AD and does not appear to be a suitable biomarker for early diagnosis of AD.
Collapse
Affiliation(s)
- Richard Novobilský
- Department of Neurology, University Hospital Ostrava, Ostrava, Czechia
- Department of Clinical Neurosciences, University of Ostrava, Ostrava, Czechia
| | - Petra Bartova
- Department of Neurology, University Hospital Ostrava, Ostrava, Czechia
- Department of Clinical Neurosciences, University of Ostrava, Ostrava, Czechia
| | - Karin Lichá
- Department of Clinical Biochemistry, Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
| | - Michal Bar
- Department of Neurology, University Hospital Ostrava, Ostrava, Czechia
- Department of Clinical Neurosciences, University of Ostrava, Ostrava, Czechia
| | - David Stejskal
- Department of Clinical Biochemistry, Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
- Institute of Laboratory Medicine, University of Ostrava, Ostrava, Czechia
| | - Pavlína Kusnierova
- Department of Clinical Biochemistry, Institute of Laboratory Medicine, University Hospital Ostrava, Ostrava, Czechia
- Institute of Laboratory Medicine, University of Ostrava, Ostrava, Czechia
| |
Collapse
|
14
|
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
|
15
|
Cocco C, Manai AL, Manca E, Noli B. Brain-Biomarker Changes in Body Fluids of Patients with Parkinson's Disease. Int J Mol Sci 2023; 24:10932. [PMID: 37446110 DOI: 10.3390/ijms241310932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disease that is rarely diagnosed at an early stage. Although the understanding of PD-related mechanisms has greatly improved over the last decade, the diagnosis of PD is still based on neurological examination through the identification of motor symptoms, including bradykinesia, rigidity, postural instability, and resting tremor. The early phase of PD is characterized by subtle symptoms with a misdiagnosis rate of approximately 16-20%. The difficulty in recognizing early PD has implications for the potential use of novel therapeutic approaches. For this reason, it is important to discover PD brain biomarkers that can indicate early dopaminergic dysfunction through their changes in body fluids, such as saliva, urine, blood, or cerebrospinal fluid (CSF). For the CFS-based test, the invasiveness of sampling is a major limitation, whereas the other body fluids are easier to obtain and could also allow population screening. Following the identification of the crucial role of alpha-synuclein (α-syn) in the pathology of PD, a very large number of studies have summarized its changes in body fluids. However, methodological problems have led to the poor diagnostic/prognostic value of this protein and alternative biomarkers are currently being investigated. The aim of this paper is therefore to summarize studies on protein biomarkers that are alternatives to α-syn, particularly those that change in nigrostriatal areas and in biofluids, with a focus on blood, and, eventually, saliva and urine.
Collapse
Affiliation(s)
- Cristina Cocco
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Antonio Luigi Manai
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Elias Manca
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
16
|
Buhmann C, Magnus T, Choe CU. Blood neurofilament light chain in Parkinson's disease. J Neural Transm (Vienna) 2023; 130:755-762. [PMID: 37067597 PMCID: PMC10199845 DOI: 10.1007/s00702-023-02632-7] [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: 02/16/2023] [Accepted: 04/01/2023] [Indexed: 04/18/2023]
Abstract
Blood neurofilament light chain (NfL) is an easily accessible, highly sensitive and reliable biomarker for neuroaxonal damage. Currently, its role in Parkinson's disease (PD) remains unclear. Here, we demonstrate that blood NfL can distinguish idiopathic PD from atypical parkinsonian syndromes (APS) with high sensitivity and specificity. In cross-sectional studies, some found significant correlations between blood NfL with motor and cognitive function, whereas others did not. In contrast, prospective studies reported very consistent associations between baseline blood NfL with motor progression and cognitive worsening. Amongst PD subtypes, especially postural instability and gait disorder (PIGD) subtype, symptoms and scores are reliably linked with blood NfL. Different non-motor PD comorbidities have also been associated with high blood NfL levels suggesting that the neuroaxonal damage of the autonomic nervous system as well as serotonergic, cholinergic and noradrenergic neurons is quantifiable. Numerous absolute NfL cutoff levels have been suggested in different cohort studies; however, validation across cohorts remains weak. However, age-adjusted percentiles and intra-individual blood NfL changes might represent more valid and consistent parameters compared with absolute NfL concentrations. In summary, blood NfL has the potential as biomarker in PD patients to be used in clinical practice for prediction of disease severity and especially progression.
Collapse
Affiliation(s)
- Carsten Buhmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Experimental Research in Stroke and Inflammation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chi-Un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Experimental Research in Stroke and Inflammation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Neurology, Klinikum Itzehoe, Robert-Koch-Straße 2, 25524, Itzehoe, Germany.
| |
Collapse
|
17
|
Gomes ESA, Van den Heuvel OA, Rietberg MB, De Groot V, Hirsch MA, Van de Berg WDJ, Jaspers RT, Vriend C, Vanbellingen T, Van Wegen EEH. (HIIT-The Track) High-Intensity Interval Training for People with Parkinson's Disease: Individual Response Patterns of (Non-)Motor Symptoms and Blood-Based Biomarkers-A Crossover Single-Case Experimental Design. Brain Sci 2023; 13:849. [PMID: 37371330 DOI: 10.3390/brainsci13060849] [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: 04/26/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION Physical exercise is receiving increasing interest as an augmentative non-pharmacological intervention in Parkinson's disease (PD). This pilot study primarily aimed to quantify individual response patterns of motor symptoms to alternating exercise modalities, along with non-motor functioning and blood biomarkers of neuroplasticity and neurodegeneration. MATERIALS & METHODS People with PD performed high-intensity interval training (HIIT) and continuous aerobic exercise (CAE) using a crossover single-case experimental design. A repeated assessment of outcome measures was conducted. The trajectories of outcome measures were visualized in time series plots and interpreted relative to the minimal clinically important difference (MCID) and smallest detectable change (SDC) or as a change in the positive or negative direction using trend lines. RESULTS Data of three participants were analyzed and engaging in physical exercise seemed beneficial for reducing motor symptoms. Participant 1 demonstrated improvement in motor function, independent of exercise modality; while for participant 2, such a clinically relevant (positive) change in motor function was only observed in response to CAE. Participant 3 showed improved motor function after HIIT, but no comparison could be made with CAE because of drop-out. Heterogeneous responses on secondary outcome measures were found, not only between exercise modalities but also among participants. CONCLUSION Though this study underpins the positive impact of physical exercise in the management of PD, large variability in individual response patterns to the interventions among participants makes it difficult to identify clear exercise-induced adaptations in functioning and blood biomarkers. Further research is needed to overcome methodological challenges in measuring individual response patterns.
Collapse
Affiliation(s)
- Elvira S Amaral Gomes
- Department of Rehabilitation Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Rehabilitation & Development, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Odile A Van den Heuvel
- Department of Anatomy & Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity & Attention, Neurodegeneration, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Marc B Rietberg
- Department of Rehabilitation Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Rehabilitation & Development, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Vincent De Groot
- Department of Rehabilitation Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Rehabilitation & Development, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neuroinfection & Neuroinflammation, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Mark A Hirsch
- Carolinas Medical Center, Atrium Health Carolinas Rehabilitation, Department of Physical Medicine and Rehabilitation, Charlotte, NC 28203, USA
- Wake Forest School of Medicine, Department of Orthopaedic Surgery and Rehabilitation, Winston-Salem, NC 27157, USA
| | - Wilma D J Van de Berg
- Department of Anatomy & Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Richard T Jaspers
- Laboratory of Myology, Department of Human Movement Science, Faculty of Behavioural and Movement Sciences, Location Vrije Universiteit Amsterdam, 1081 BT Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Rehabilitation & Development, Tissue Function & Regeneration, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Chris Vriend
- Department of Anatomy & Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity & Attention, Neurodegeneration, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Tim Vanbellingen
- Gerontechnology and Rehabilitation Group, University of Bern, 3008 Bern, Switzerland
- Neurocenter, Luzerner Kantonsspital, 6000 Lucerne, Switzerland
| | - Erwin E H Van Wegen
- Department of Rehabilitation Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Rehabilitation & Development, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Ageing & Vitality, Amsterdam UMC, Location Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam UMC, Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
18
|
Zhang X, Ma L, Liang D, Song B, Chen J, Huang Y, Xu L, Zhao P, Wu W, Zhang N, Xue R. Neurofilament Light Protein Predicts Disease Progression in Idiopathic REM Sleep Behavior Disorder. JOURNAL OF PARKINSON'S DISEASE 2023:JPD223519. [PMID: 37182898 DOI: 10.3233/jpd-223519] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Idiopathic rapid eye movement sleep behavior disorder (iRBD) is increasingly recognized as a manifestation preceding the α-synucleinopathies like Parkinson's disease (PD). Neurofilament light chain (NfL) have been reported to be higher in synucleinopathies as a sign of neurodegeneration. OBJECTIVE To evaluate whether plasma NfL is valuable in reflecting cognitive and motor status in iRBD and PD with a premorbid history of RBD (PDRBD), and predicting disease progression in iRBD. METHODS Thirty-one patients with iRBD, 30 with PDRBD, and 18 healthy controls were included in the cross-sectional and prospective study. Another cohort from the Parkinson's Progression Markers Initiative (PPMI) dataset was enrolled for verification analysis. All patients received evaluations of cognitive, motor, and autonomic function by a battery of clinical tests at baseline and follow-up. Blood NfL was measured by the Quanterix Simoa HD-1. RESULTS In our cohort, 26 patients with iRBD completed the follow-up evaluations, among whom eight (30.8%) patients displayed phenoconversion. Baseline plasma NfL cutoff value of 22.93 pg/mL performed best in distinguishing the iRBD converters from non-converters (sensitivity: 75.0%, specificity: 83.3%, area under the curve: 0.84). Cognitive and motor function were significantly correlated with NfL levels in PDRBD (correlation coefficients: -0.379, 0.399; respectively). Higher baseline NfL levels in iRBD were significantly associated with higher risks for cognitive, motor, autonomic function progression, and phenoconversion at follow-up (hazard ratios: 1.069, 1.065, 1.170, 1.065; respectively). The findings were supported by the PPMI dataset. CONCLUSION Plasma NfL is valuable in reflecting disease severity of PDRBD and predicting disease progression and phenoconversion in iRBD.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Li Ma
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Danqi Liang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingxin Song
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingshan Chen
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaqin Huang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Wu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Xue
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Mao S, Teng X, Li Z, Zu J, Zhang T, Xu C, Cui G. Association of serum neurofilament light chain and glial fibrillary acidic protein levels with cognitive decline in Parkinson's disease. Brain Res 2023; 1805:148271. [PMID: 36754139 DOI: 10.1016/j.brainres.2023.148271] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVES To investigate whether serum neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) levels are associated with motor and cognitive function in Parkinson's disease (PD). METHODS This cross-sectional study recruited 140 participants, including 103 PD patients and 37 healthy controls (HC). Serum NfL and GFAP levels were measured using the ultrasensitive single-molecule array (Simoa) technique. Motor and cognitive function were evaluated using the Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III (MDS-UPDRS III) and Beijing version of the Montreal Cognitive Assessment (MoCA). Spearman's correlation analyses were used to determine the correlation between serum NfL and GFAP levels and clinical features in PD patients. Binary logistic regression analysis was used to assess the association between serum biomarkers and cognitive impairment in PD patients. RESULTS We observed significantly higher serum NfL and GFAP levels in PD patients than in HC (p < 0.001). Serum NfL and GFAP levels were negatively correlated with MoCA scores (NfL: r = - 0.472, p < 0.001; r = 0.395, p < 0.001) and multiple cognitive domains and showed no correlation with motor symptom severity after adjusting for age and sex. Binary logistic regression analysis showed that the serum NfL and GFAP levels were independent contributors to PD with dementia (p < 0.05). CONCLUSIONS Both serum NfL and GFAP levels correlated with cognitive impairment, but not motor symptoms, in PD patients. Serum NfL and GFAP levels can serve as biomarkers for PD patients at risk of cognitive decline.
Collapse
Affiliation(s)
- Shuai Mao
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Xing Teng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Zhen Li
- Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China
| | - Tao Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China
| | - Chuanying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China.
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China.
| |
Collapse
|
20
|
Youssef P, Hughes L, Kim WS, Halliday GM, Lewis SJG, Cooper A, Dzamko N. Evaluation of plasma levels of NFL, GFAP, UCHL1 and tau as Parkinson's disease biomarkers using multiplexed single molecule counting. Sci Rep 2023; 13:5217. [PMID: 36997567 PMCID: PMC10063670 DOI: 10.1038/s41598-023-32480-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 03/28/2023] [Indexed: 04/01/2023] Open
Abstract
Objective biomarkers for Parkinson's Disease (PD) could aid early and specific diagnosis, effective monitoring of disease progression, and improved design and interpretation of clinical trials. Although alpha-synuclein remains a biomarker candidate of interest, the multifactorial and heterogenous nature of PD highlights the need for a PD biomarker panel. Ideal biomarker candidates include markers that are detectable in easily accessible samples, (ideally blood) and that reflect the underlying pathological process of PD. In the present study, we explored the diagnostic and prognostic PD biomarker potential of the SIMOA neurology 4-plex-A biomarker panel, which included neurofilament light (NFL), glial fibrillary acid protein (GFAP), tau and ubiquitin C-terminal hydrolase L1 (UCHL-1). We initially performed a serum vs plasma comparative study to determine the most suitable blood-based matrix for the measurement of these proteins in a multiplexed assay. The levels of NFL and GFAP in plasma and serum were highly correlated (Spearman rho-0.923, p < 0.0001 and rho = 0.825, p < 0.001 respectively). In contrast, the levels of tau were significantly higher in plasma compared to serum samples (p < 0.0001) with no correlation between sample type (Spearman p > 0.05). The neurology 4-plex-A panel, along with plasma alpha-synuclein was then assessed in a cross-sectional cohort of 29 PD patients and 30 controls. Plasma NFL levels positively correlated with both GFAP and alpha-synuclein levels (rho = 0.721, p < 0.0001 and rho = 0.390, p < 0.05 respectively). As diagnostic biomarkers, the control and PD groups did not differ in their mean NFL, GFAP, tau or UCHL-1 plasma levels (t test p > 0.05). As disease state biomarkers, motor severity (MDS-UPDRS III) correlated with increased NFL (rho = 0.646, p < 0.0001), GFAP (rho = 0.450, p < 0.05) and alpha-synuclein levels (rho = 0.406, p < 0.05), while motor stage (Hoehn and Yahr) correlated with increased NFL (rho = 0.455, p < 0.05) and GFAP (rho = 0.549, p < 0.01) but not alpha-synuclein levels (p > 0.05). In conclusion, plasma was determined to be most suitable blood-based matrix for multiplexing the neurology 4-plex-A panel. Given their correlation with motor features of PD, NFL and GFAP appear to be promising disease state biomarker candidates and further longitudinal validation of these two proteins as blood-based biomarkers for PD progression is warranted.
Collapse
Affiliation(s)
- Priscilla Youssef
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Laura Hughes
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Woojin S Kim
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Glenda M Halliday
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Simon J G Lewis
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Antony Cooper
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, UNSW-Sydney, Darlinghurst, NSW, 2010, Australia
| | - Nicolas Dzamko
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
21
|
Canever JB, Soares ES, de Avelar NCP, Cimarosti HI. Targeting α-synuclein post-translational modifications in Parkinson's disease. Behav Brain Res 2023; 439:114204. [PMID: 36372243 DOI: 10.1016/j.bbr.2022.114204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the nigrostriatal pathway. Although the exact mechanisms underlying PD are still not completely understood, it is well accepted that α-synuclein plays key pathophysiological roles as the main constituent of the cytoplasmic inclusions known as Lewy bodies. Several post-translational modifications (PTMs), such as the best-known phosphorylation, target α-synuclein and are thus implicated in its physiological and pathological functions. In this review, we present (1) an overview of the pathophysiological roles of α-synuclein, (2) a descriptive analysis of α-synuclein PTMs, including phosphorylation, ubiquitination, SUMOylation, acetylation, glycation, truncation, and O-GlcNAcylation, as well as (3) a brief summary on α-synuclein PTMs as potential biomarkers for PD. A better understanding of α-synuclein PTMs is of paramount importance for elucidating the mechanisms underlying PD and can thus be expected to improve early detection and monitoring disease progression, as well as identify promising new therapeutic targets.
Collapse
Affiliation(s)
- Jaquelini B Canever
- Post-Graduate Program in Neuroscience, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Laboratory of Aging, Resources and Rheumatology, UFSC, Araranguá, Santa Catarina, Brazil
| | - Ericks Sousa Soares
- Post-Graduate Program in Pharmacology, UFSC, Florianópolis, Santa Catarina, Brazil
| | - Núbia C P de Avelar
- Laboratory of Aging, Resources and Rheumatology, UFSC, Araranguá, Santa Catarina, Brazil
| | - Helena I Cimarosti
- Post-Graduate Program in Neuroscience, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Post-Graduate Program in Pharmacology, UFSC, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
22
|
Abdi IY, Bartl M, Dakna M, Abdesselem H, Majbour N, Trenkwalder C, El-Agnaf O, Mollenhauer B. Cross-sectional proteomic expression in Parkinson's disease-related proteins in drug-naïve patients vs healthy controls with longitudinal clinical follow-up. Neurobiol Dis 2023; 177:105997. [PMID: 36634823 DOI: 10.1016/j.nbd.2023.105997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
There is an urgent need to find reliable and accessible blood-based biomarkers for early diagnosis of Parkinson's disease (PD) correlating with clinical symptoms and displaying predictive potential to improve future clinical trials. This led us to a conduct large-scale proteomics approach using an advanced high-throughput proteomics technology to create a proteomic profile for PD. Over 1300 proteins were measured in serum samples from a de novo Parkinson's (DeNoPa) cohort made up of 85 deep clinically phenotyped drug-naïve de novo PD patients and 93 matched healthy controls (HC) with longitudinal clinical follow-up available of up to 8 years. The analysis identified 73 differentially expressed proteins (DEPs) of which 14 proteins were confirmed as stable potential diagnostic markers using machine learning tools. Among the DEPs identified, eight proteins-ALCAM, contactin 1, CD36, DUS3, NEGR1, Notch1, TrkB, and BTK- significantly correlated with longitudinal clinical scores including motor and non-motor symptom scores, cognitive function and depression scales, indicating potential predictive values for progression in PD among various phenotypes. Known functions of these proteins and their possible relation to the pathophysiology or symptomatology of PD were discussed and presented with a particular emphasis on the potential biological mechanisms involved, such as cell adhesion, axonal guidance and neuroinflammation, and T-cell activation. In conclusion, with the use of advance multiplex proteomic technology, a blood-based protein signature profile was identified from serum samples of a well-characterized PD cohort capable of potentially differentiating PD from HC and predicting clinical disease progression of related motor and non-motor PD symptoms. We thereby highlight the need to validate and further investigate these markers in future prospective cohorts and assess their possible PD-related mechanisms.
Collapse
Affiliation(s)
- Ilham Yahya Abdi
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar; Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Houari Abdesselem
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Nour Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Klinikstrasse, Kassel, Germany; Department of Neurosurgery, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Omar El-Agnaf
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar; Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany; Paracelsus-Elena-Klinik, Klinikstrasse, Kassel, Germany.
| |
Collapse
|
23
|
Frigerio I, Laansma MA, Lin CP, Hermans EJM, Bouwman MMA, Bol JGJM, Galis-de Graaf Y, Hepp DH, Rozemuller AJM, Barkhof F, van de Berg WDJ, Jonkman LE. Neurofilament light chain is increased in the parahippocampal cortex and associates with pathological hallmarks in Parkinson's disease dementia. Transl Neurodegener 2023; 12:3. [PMID: 36658627 PMCID: PMC9854202 DOI: 10.1186/s40035-022-00328-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/17/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Increased neurofilament levels in biofluids are commonly used as a proxy for neurodegeneration in several neurodegenerative disorders. In this study, we aimed to investigate the distribution of neurofilaments in the cerebral cortex of Parkinson's disease (PD), PD with dementia (PDD) and dementia with Lewy bodies (DLB) donors, and its association with pathology load and MRI measures of atrophy and diffusivity. METHODS Using a within-subject post-mortem MRI-pathology approach, we included 9 PD, 12 PDD/DLB and 18 age-matched control donors. Cortical thickness and mean diffusivity (MD) metrics were extracted respectively from 3DT1 and DTI at 3T in-situ MRI. After autopsy, pathological hallmarks (pSer129-αSyn, p-tau and amyloid-β load) together with neurofilament light-chain (NfL) and phosphorylated-neurofilament medium- and heavy-chain (p-NfM/H) immunoreactivity were quantified in seven cortical regions, and studied in detail with confocal-laser scanning microscopy. The correlations between MRI and pathological measures were studied using linear mixed models. RESULTS Compared to controls, p-NfM/H immunoreactivity was increased in all cortical regions in PD and PDD/DLB, whereas NfL immunoreactivity was increased in the parahippocampal and entorhinal cortex in PDD/DLB. NfL-positive neurons showed degenerative morphological features and axonal fragmentation. The increased p-NfM/H correlated with p-tau load, and NfL correlated with pSer129-αSyn but more strongly with p-tau load in PDD/DLB. Lastly, neurofilament immunoreactivity correlated with cortical thinning in PD and with increased cortical MD in PDD/DLB. CONCLUSIONS Taken together, increased neurofilament immunoreactivity suggests underlying axonal injury and neurofilament accumulation in morphologically altered neurons with increased pathological burden. Importantly, we demonstrate that such neurofilament markers at least partly explain MRI measures that are associated with the neurodegenerative process.
Collapse
Affiliation(s)
- Irene Frigerio
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands. .,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands. .,Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands.
| | - Max A. Laansma
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Chen-Pei Lin
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Emma J. M. Hermans
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Maud M. A. Bouwman
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - John G. J. M. Bol
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Yvon Galis-de Graaf
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Dagmar H. Hepp
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Neurology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Annemieke J. M. Rozemuller
- grid.12380.380000 0004 1754 9227Department of Pathology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Frederik Barkhof
- grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - Wilma D. J. van de Berg
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Laura E. Jonkman
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Endoplasmic Reticulum Stress-Regulated Chaperones as a Serum Biomarker Panel for Parkinson's Disease. Mol Neurobiol 2023; 60:1476-1485. [PMID: 36478320 PMCID: PMC9899193 DOI: 10.1007/s12035-022-03139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Examination of post-mortem brain tissues has previously revealed a strong association between Parkinson's disease (PD) pathophysiology and endoplasmic reticulum (ER) stress. Evidence in the literature regarding the circulation of ER stress-regulated factors released from neurons provides a rationale for investigating ER stress biomarkers in the blood to aid diagnosis of PD. The levels of ER stress-regulated proteins in serum collected from 29 PD patients and 24 non-PD controls were measured using enzyme-linked immunosorbent assays. A panel of four biomarkers, protein disulfide-isomerase A1, protein disulfide-isomerase A3, mesencephalic astrocyte-derived neurotrophic factor, and clusterin, together with age and gender had higher ability (area under the curve 0.64, sensitivity 66%, specificity 57%) and net benefit to discriminate PD patients from the non-PD group compared with other analyzed models. Addition of oligomeric and total α-synuclein to the model did not improve the diagnostic power of the biomarker panel. We provide evidence that ER stress-regulated proteins merit further investigation for their potential as diagnostic biomarkers of PD.
Collapse
|
25
|
Gonzalez-Robles C, Weil RS, van Wamelen D, Bartlett M, Burnell M, Clarke CS, Hu MT, Huxford B, Jha A, Lambert C, Lawton M, Mills G, Noyce A, Piccini P, Pushparatnam K, Rochester L, Siu C, Williams-Gray CH, Zeissler ML, Zetterberg H, Carroll CB, Foltynie T, Schrag A. Outcome Measures for Disease-Modifying Trials in Parkinson's Disease: Consensus Paper by the EJS ACT-PD Multi-Arm Multi-Stage Trial Initiative. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1011-1033. [PMID: 37545260 PMCID: PMC10578294 DOI: 10.3233/jpd-230051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/23/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Multi-arm, multi-stage (MAMS) platform trials can accelerate the identification of disease-modifying treatments for Parkinson's disease (PD) but there is no current consensus on the optimal outcome measures (OM) for this approach. OBJECTIVE To provide an up-to-date inventory of OM for disease-modifying PD trials, and a framework for future selection of OM for such trials. METHODS As part of the Edmond J Safra Accelerating Clinical Trials in Parkinson Disease (EJS ACT-PD) initiative, an expert group with Patient and Public Involvement and Engagement (PPIE) representatives' input reviewed and evaluated available evidence on OM for potential use in trials to delay progression of PD. Each OM was ranked based on aspects such as validity, sensitivity to change, participant burden and practicality for a multi-site trial. Review of evidence and expert opinion led to the present inventory. RESULTS An extensive inventory of OM was created, divided into: general, motor and non-motor scales, diaries and fluctuation questionnaires, cognitive, disability and health-related quality of life, capability, quantitative motor, wearable and digital, combined, resource use, imaging and wet biomarkers, and milestone-based. A framework for evaluation of OM is presented to update the inventory in the future. PPIE input highlighted the need for OM which reflect their experience of disease progression and are applicable to diverse populations and disease stages. CONCLUSION We present a range of OM, classified according to a transparent framework, to aid selection of OM for disease-modifying PD trials, whilst allowing for inclusion or re-classification of relevant OM as new evidence emerges.
Collapse
Affiliation(s)
| | | | | | | | - Matthew Burnell
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Li Y, Zheng JJ, Wu X, Gao W, Liu CJ. Postural control of Parkinson's disease: A visualized analysis based on Citespace knowledge graph. Front Aging Neurosci 2023; 15:1136177. [PMID: 37032828 PMCID: PMC10080997 DOI: 10.3389/fnagi.2023.1136177] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Postural control impairment is one of the primary motor symptoms in patients with Parkinson's disease, leading to an increased risk of falling. Several studies have been conducted on postural control disorders in Parkinson's disease patients, but no relevant bibliometric analysis has been found. In this paper, the Web of Science Core Collection database was searched for 1,295 relevant papers on postural control in Parkinson's disease patients from December 2011 to December 2021. Based on the Citespace knowledge graph, these relevant papers over the last decade were analyzed from the perspectives of annual publication volume, countries and institutes cooperation, authors cooperation, dual-map overlay of journals, co-citation literature, and keywords. The purpose of this study was to explore the current research status, research hotspots, and frontiers in this field, and to provide a reference for further promoting the research on postural control in Parkinson's disease patients.
Collapse
Affiliation(s)
- Yan Li
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Jie-Jiao Zheng
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Shanghai Clinical Research Center for Rehabilitation Medicine, Shanghai, China
- *Correspondence: Jie-Jiao Zheng,
| | - Xie Wu
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Wen Gao
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Rehabilitation Medicine, Shanghai, China
| | - Chan-Jing Liu
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
27
|
A meta-analysis of the diagnostic utility of biomarkers in cerebrospinal fluid in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:165. [PMID: 36446820 PMCID: PMC9709054 DOI: 10.1038/s41531-022-00431-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Biomarkers play important roles in the diagnosis and differential diagnosis of Parkinson's disease (PD). Thus, we carried out a systematic review and meta-analysis evaluating the diagnostic utility of cerebrospinal fluid (CSF) biomarkers to distinguish PD from atypical parkinsonian syndromes (APSs) and controls. Data for PD and APS and controls were extracted from 123 studies that reported the concentration of CSF biomarkers. Comparisons were presented using pooled Hedges' g. Sources of heterogeneity were evaluated using meta-regression, and subgroup and sensitivity analyses. We found that compared with controls, PD patients had lower levels of amyloid beta 1-42, phosphorylated tau, total tau, total α-synuclein, Zn, DJ-1, and YKL-40, and higher levels of oligomeric and phosphorylated α-synuclein. Moreover, lower CSF levels of neurofilament light chain, t-tau, YKL-40, and C-reactive protein were found in PD patients compared to those with multiple system atrophy. PD patients also had lower levels of NFL and higher levels of Aβ42 compared with patients with progressive supranuclear palsy. Reduced levels of p-tau and t-tau and higher Aβ42 levels were found in PD patients compared with patients with dementia with Lewy bodies. Finally, reduced NFL levels were found in patients with PD compared with patients with cortical basal degeneration. Therefore, we believe that the combinations of t-α-syn, Aβ42, and NFL could be promising biomarkers for the differential diagnosis of PD and APSs.
Collapse
|
28
|
Vermunt L, Otte M, Verberk IMW, Killestein J, Lemstra AW, van der Flier WM, Pijnenburg YAL, Vijverberg EGB, Bouwman FH, Gravesteijn G, van de Berg WDJ, Scheltens P, van Harten AC, Willemse EAJ, Teunissen CE. Age- and disease-specific reference values for neurofilament light presented in an online interactive support interface. Ann Clin Transl Neurol 2022; 9:1832-1837. [PMID: 36196979 DOI: 10.1002/acn3.51676] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Interpretation of axonal damage biomarker Neurofilament Light chain (NfL) concentrations is difficult due to the lack of age-specific and disease-specific reference values. We here developed an interactive interface to support interpretation of NfL results in human body fluids. We used NfL values of 1698 individuals without a neurological disorder, aged 19-85 years, and patients with MS and dementias. Percentile regression estimates per diagnosis populate interactive graphs, alongside NfL background information (available on: https://mybiomarkers.shinyapps.io/Neurofilament). This accessible interface provides reference for interpretation of the individual patient results for clinicians. It showcases an adaptable method to support interpretation of age-dependent biomarkers in neurology.
Collapse
Affiliation(s)
- Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marco Otte
- Network Institute Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joep Killestein
- MS Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Everard G B Vijverberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Femke H Bouwman
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gido Gravesteijn
- Department of Clinical Genetics and Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wilma D J van de Berg
- Clinical Neuroanatomy and Biobanking, department of Anatomy and Neurosciences, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Life Science Partners (LSP), Amsterdam, The Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Eline A J Willemse
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Liu Y, Dou K, Xue L, Li X, Xie A. Neurofilament light as a biomarker for motor decline in Parkinson’s disease. Front Neurosci 2022; 16:959261. [PMID: 36117629 PMCID: PMC9477093 DOI: 10.3389/fnins.2022.959261] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/05/2022] [Indexed: 11/15/2022] Open
Abstract
Objectives The aim of this study was to determine whether neurofifilament light (NfL) could reflect motor decline and compare the predictive values of cerebrospinal fluid (CSF) and serum NfL in individuals with PD. Methods CSF/serum samples were collected from patients with PD and healthy controls (HCs) with motor assessments at baseline and after three years of follow-up from the Parkinson’s Progression Markers Initiative (PPMI). Multiple linear regression models and linear mixed-effects models were used to investigate the associations of motor assessments with baseline and longitudinal CSF/serum NfL. Associations between the change rates of motor assessments and CSF/serum NfL were further investigated via multiple linear regression models. Mediating effect analysis was used to research whether CSF alpha-synuclein (α-syn) acts as the mediator between NfL and motor assessments. Results We found patients with PD had higher baseline CSF/serum NfL levels than HCs. Both baseline CSF/serum NfLs and their change rates predicted measurable motor decline in PD assessed by different motor scores. Baseline serum NfL and its rate of change were strongly associated with CSF NfL levels in patients with PD (P < 0.001). Besides, there were also significant differences in CSF/serum NfL levels and predicted values of motor decline between men and women with PD. Mediating effect analysis showed CSF α-syn mediated the effect of CSF NfL on total Unified Parkinson’s Disease Rating Scale (UPDRS) scores and UPDRSIII with 30.6 and 20.2% mediation, respectively. Conclusion Our results indicated that NfL, especially serum NfL concentration, could serve as an easily accessible biomarker to monitor the severity and progression of motor decline in individuals with PD, especially in men with PD. Besides, CSF α-syn acts as a mediator between NfL and motor progression.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kaixin Dou
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ling Xue
- Department of Nursing, Tai’an City Central Hospital, Tai’an, China
| | - Xiaoyuan Li
- Department of Traditional Chinese Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Anmu Xie,
| |
Collapse
|
30
|
Tönges L, Buhmann C, Klebe S, Klucken J, Kwon EH, Müller T, Pedrosa DJ, Schröter N, Riederer P, Lingor P. Blood-based biomarker in Parkinson's disease: potential for future applications in clinical research and practice. J Neural Transm (Vienna) 2022; 129:1201-1217. [PMID: 35428925 PMCID: PMC9463345 DOI: 10.1007/s00702-022-02498-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/27/2022] [Indexed: 12/12/2022]
Abstract
The clinical presentation of Parkinson's disease (PD) is both complex and heterogeneous, and its precise classification often requires an intensive work-up. The differential diagnosis, assessment of disease progression, evaluation of therapeutic responses, or identification of PD subtypes frequently remains uncertain from a clinical point of view. Various tissue- and fluid-based biomarkers are currently being investigated to improve the description of PD. From a clinician's perspective, signatures from blood that are relatively easy to obtain would have great potential for use in clinical practice if they fulfill the necessary requirements as PD biomarker. In this review article, we summarize the knowledge on blood-based PD biomarkers and present both a researcher's and a clinician's perspective on recent developments and potential future applications.
Collapse
Affiliation(s)
- Lars Tönges
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany.
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, 44801, Bochum, Nordrhein-Westfalen, Germany.
| | - Carsten Buhmann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Stephan Klebe
- Department of Neurology, University Hospital Essen, 45147, Essen, Germany
| | - Jochen Klucken
- Department of Digital Medicine, University Luxembourg, LCSB, L-4367, Belval, Luxembourg
- Digital Medicine Research Group, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Centre Hospitalier de Luxembourg, Digital Medicine Research Clinic, L-1210, Luxembourg, Luxembourg
| | - Eun Hae Kwon
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Gudrunstr. 56, 44791, Bochum, Germany
| | - Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weissensee, 13088, Berlin, Germany
| | - David J Pedrosa
- Department of Neurology, Universitätsklinikum Gießen and Marburg, Marburg Site, 35043, Marburg, Germany
- Center of Mind, Brain and Behaviour (CMBB), Philipps-Universität Marburg, 35043, Marburg, Germany
| | - Nils Schröter
- Department of Neurology and Clinical Neuroscience, University of Freiburg, 79106, Freiburg, Germany
| | - Peter Riederer
- Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, 97080, Wuerzburg, Germany
- University of Southern Denmark Odense, 5000, Odense, Denmark
| | - Paul Lingor
- School of Medicine, Klinikum Rechts Der Isar, Department of Neurology, Technical University of Munich, 81675, München, Germany
| |
Collapse
|
31
|
Dou K, Ma J, Zhang X, Shi W, Tao M, Xie A. Multi-predictor modeling for predicting early Parkinson’s disease and non-motor symptoms progression. Front Aging Neurosci 2022; 14:977985. [PMID: 36092799 PMCID: PMC9459236 DOI: 10.3389/fnagi.2022.977985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Identifying individuals with high-risk Parkinson’s disease (PD) at earlier stages is an urgent priority to delay disease onset and progression. In the present study, we aimed to develop and validate clinical risk models using non-motor predictors to distinguish between early PD and healthy individuals. In addition, we constructed prognostic models for predicting the progression of non-motor symptoms [cognitive impairment, Rapid-eye-movement sleep Behavior Disorder (RBD), and depression] in de novo PD patients at 5 years of follow-up. Methods We retrieved the data from the Parkinson’s Progression Markers Initiative (PPMI) database. After a backward variable selection approach to identify predictors, logistic regression analyses were applied for diagnosis model construction, and cox proportional-hazards models were used to predict non-motor symptom progression. The predictive models were internally validated by correcting measures of predictive performance for “optimism” or overfitting with the bootstrap resampling approach. Results For constructing diagnostic models, the final model reached a high accuracy with an area under the curve (AUC) of 0.93 (95% CI: 0.91–0.96), which included eight variables (age, gender, family history, University of Pennsylvania Smell Inventory Test score, Montreal Cognitive Assessment score, RBD Screening Questionnaire score, levels of cerebrospinal fluid α-synuclein, and SNCA rs356181 polymorphism). For the construction of prognostic models, our results showed that the AUC of the three prognostic models improved slightly with increasing follow-up time. The overall AUCs fluctuated around 0.70. The model validation established good discrimination and calibration for predicting PD onset and progression of non-motor symptoms. Conclusion The findings of our study facilitate predicting the individual risk at an early stage based on the predictors derived from these models. These predictive models provide relatively reliable information to prevent PD onset and progression. However, future validation analysis is still needed to clarify these findings and provide more insight into the predictive models over more extended periods of disease progression in more diverse samples.
Collapse
|
32
|
Altered BDNF levels are associated with cognitive impairment in Parkinson's disease patients with depression. Parkinsonism Relat Disord 2022; 103:122-128. [DOI: 10.1016/j.parkreldis.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022]
|
33
|
Opportunities and challenges of alpha-synuclein as a potential biomarker for Parkinson's disease and other synucleinopathies. NPJ Parkinsons Dis 2022; 8:93. [PMID: 35869066 PMCID: PMC9307631 DOI: 10.1038/s41531-022-00357-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD), the second most common progressive neurodegenerative disease, develops and progresses for 10–15 years before the clinical diagnostic symptoms of the disease are manifested. Furthermore, several aspects of PD pathology overlap with other neurodegenerative diseases (NDDs) linked to alpha-synuclein (aSyn) aggregation, also called synucleinopathies. Therefore, there is an urgent need to discover and validate early diagnostic and prognostic markers that reflect disease pathophysiology, progression, severity, and potential differences in disease mechanisms between PD and other NDDs. The close association between aSyn and the development of pathology in synucleinopathies, along with the identification of aSyn species in biological fluids, has led to increasing interest in aSyn species as potential biomarkers for early diagnosis of PD and differentiate it from other synucleinopathies. In this review, we (1) provide an overview of the progress toward mapping the distribution of aSyn species in the brain, peripheral tissues, and biological fluids; (2) present comparative and critical analysis of previous studies that measured total aSyn as well as other species such as modified and aggregated forms of aSyn in different biological fluids; and (3) highlight conceptual and technical gaps and challenges that could hinder the development and validation of reliable aSyn biomarkers; and (4) outline a series of recommendations to address these challenges. Finally, we propose a combined biomarker approach based on integrating biochemical, aggregation and structure features of aSyn, in addition to other biomarkers of neurodegeneration. We believe that capturing the diversity of aSyn species is essential to develop robust assays and diagnostics for early detection, patient stratification, monitoring of disease progression, and differentiation between synucleinopathies. This could transform clinical trial design and implementation, accelerate the development of new therapies, and improve clinical decisions and treatment strategies.
Collapse
|
34
|
Wong YY, Wu CY, Yu D, Kim E, Wong M, Elez R, Zebarth J, Ouk M, Tan J, Liao J, Haydarian E, Li S, Fang Y, Li P, Pakosh M, Tartaglia MC, Masellis M, Swardfager W. Biofluid markers of blood-brain barrier disruption and neurodegeneration in Lewy body spectrum diseases: A systematic review and meta-analysis. Parkinsonism Relat Disord 2022; 101:119-128. [PMID: 35760718 DOI: 10.1016/j.parkreldis.2022.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mixed evidence supports blood-brain barrier (BBB) dysfunction in Lewy body spectrum diseases. METHODS We compare biofluid markers in people with idiopathic Parkinson's disease (PD) and people with PD dementia (PDD) and/or dementia with Lewy bodies (DLB), compared with healthy controls (HC). Seven databases were searched up to May 10, 2021. Outcomes included cerebrospinal fluid to blood albumin ratio (Qalb), and concentrations of 7 blood protein markers that also reflect BBB disruption and/or neurodegenerative co-pathology. We further explore differences between PD patients with and without evidence of dementia. Random-effects models were used to obtain standardized mean differences (SMD) with 95% confidence interval. RESULTS Of 13,949 unique records, 51 studies were meta-analyzed. Compared to HC, Qalb was higher in PD (NPD/NHC = 224/563; SMD = 0.960 [0.227-1.694], p = 0.010; I2 = 92.2%) and in PDD/DLB (NPDD/DLB/NHC = 265/670; SMD = 1.126 [0.358-1.893], p < 0.001; I2 = 78.2%). Blood neurofilament light chain (NfL) was higher in PD (NPD/NHC = 1848/1130; SMD = 0.747 [0.442-1.052], p < 0.001; I2 = 91.9%) and PDD/DLB (NPDD/DLB/NHC = 183/469; SMD = 1.051 [0.678-1.423], p = 0.004; I2 = 92.7%) than in HC. p-tau 181 (NPD/NHC = 276/164; SMD = 0.698 [0.149-1.247], p = 0.013; I2 = 82.7%) was also higher in PD compared to HC. In exploratory analyses, blood NfL was higher in PD without dementia (NPDND/NHC = 1005/740; SMD = 0.252 [0.042-0.462], p = 0.018; I2 = 71.8%) and higher in PDD (NPDD/NHC = 100/111; SMD = 0.780 [0.347-1.214], p < 0.001; I2 = 46.7%) compared to HC. Qalb (NPDD/NPDND = 63/191; SMD = 0.482 [0.189-0.774], p = 0.010; I2<0.001%) and NfL (NPDD/NPDND = 100/223; SMD = 0.595 [0.346-0.844], p < 0.001; I2 = 3.4%) were higher in PDD than in PD without dementia. CONCLUSIONS Biofluid markers suggest BBB disruption and neurodegenerative co-pathology involvement in common Lewy body diseases. Greater evidence of BBB breakdown was seen in Lewy body disease with cognitive impairment.
Collapse
Affiliation(s)
- Yuen Yan Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Di Yu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Esther Kim
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Renata Elez
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Zebarth
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Ouk
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jocelyn Tan
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jiamin Liao
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Eileen Haydarian
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Siming Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Yaolu Fang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Peihao Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Maureen Pakosh
- Library & Information Services, UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Mario Masellis
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Zubelzu M, Morera-Herreras T, Irastorza G, Gómez-Esteban JC, Murueta-Goyena A. Plasma and serum alpha-synuclein as a biomarker in Parkinson's disease: A meta-analysis. Parkinsonism Relat Disord 2022; 99:107-115. [PMID: 35717321 DOI: 10.1016/j.parkreldis.2022.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Reliable biomarkers for Parkinson's disease (PD) diagnosis are urgently needed. Alpha-synuclein (α-syn) and its proteoforms play a key role in PD pathology but in vivo measurements have raised conflicting results, and whether α-syn in blood could distinguish PD patients from healthy controls is still controversial. METHODS A systematic literature search yielded 35 eligible studies for meta-analysis reporting the concentration of total, oligomeric or phosphorylated α-syn in plasma and/or serum of PD patients and healthy controls. Standardized mean differences (SMD) were pooled using multivariate/multilevel linear mixed-effects models. Meta-regression analyses were conducted to investigate possible modifiers. RESULTS A meta-analysis of 32 articles involving 2683 PD patients and 1838 controls showed a significant overall effect of PD on total α-syn levels (SMD = 0.85, p = 0.004). Meta-regression showed that increased SMD of total α-syn in PD was significantly associated with lower age, shorter disease duration, mild motor impairment, and Immunomagnetic Reduction assay for protein quantification. In contrast, no significant differences were observed for oligomeric or phosphorylated α-syn between PD and controls but increased oligomeric α-syn was significantly associated with shorter disease duration. The heterogeneity among studies was high (>98%). CONCLUSIONS These findings suggest that increased total plasma/serum α-syn levels in PD primarily occur in early phases of the disease. The evidence obtained from a small number of studies measuring plasma/serum concentrations of oligomeric and phosphorylated species of α-syn shows no difference. The clinical applicability of measuring plasma or serum α-syn species for differentiating PD from healthy control warrants further studies with better clinical profiling of PD patients.
Collapse
Affiliation(s)
- Maider Zubelzu
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| | - Gorka Irastorza
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Juan Carlos Gómez-Esteban
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain; Department of Neurology, Cruces University Hospital, Osakidetza, Barakaldo, Bizkaia, Spain
| | - Ane Murueta-Goyena
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain; Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
36
|
Vijiaratnam N, Lawton M, Heslegrave AJ, Guo T, Tan M, Jabbari E, Real R, Woodside J, Grosset K, Chelban V, Athauda D, Girges C, Barker RA, Hardy J, Wood N, Houlden H, Williams N, Ben-Shlomo Y, Zetterberg H, Grosset DG, Foltynie T, Morris HR. Combining biomarkers for prognostic modelling of Parkinson's disease. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328365. [PMID: 35577512 PMCID: PMC9279845 DOI: 10.1136/jnnp-2021-328365] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patients with Parkinson's disease (PD) have variable rates of progression. More accurate prediction of progression could improve selection for clinical trials. Although some variance in clinical progression can be predicted by age at onset and phenotype, we hypothesise that this can be further improved by blood biomarkers. OBJECTIVE To determine if blood biomarkers (serum neurofilament light (NfL) and genetic status (glucocerebrosidase, GBA and apolipoprotein E (APOE))) are useful in addition to clinical measures for prognostic modelling in PD. METHODS We evaluated the relationship between serum NfL and baseline and longitudinal clinical measures as well as patients' genetic (GBA and APOE) status. We classified patients as having a favourable or an unfavourable outcome based on a previously validated model, and explored how blood biomarkers compared with clinical variables in distinguishing prognostic phenotypes . RESULTS 291 patients were assessed in this study. Baseline serum NfL was associated with baseline cognitive status. Nfl predicted a shorter time to dementia, postural instability and death (dementia-HR 2.64; postural instability-HR 1.32; mortality-HR 1.89) whereas APOEe4 status was associated with progression to dementia (dementia-HR 3.12, 95% CI 1.63 to 6.00). NfL levels and genetic variables predicted unfavourable progression to a similar extent as clinical predictors. The combination of clinical, NfL and genetic data produced a stronger prediction of unfavourable outcomes compared with age and gender (area under the curve: 0.74-age/gender vs 0.84-ALL p=0.0103). CONCLUSIONS Clinical trials of disease-modifying therapies might usefully stratify patients using clinical, genetic and NfL status at the time of recruitment.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Michael Lawton
- Population Health Sciences, University of Bristol, Bristol, UK
- Department of Social Medicine, University of Bristol, Bristol, UK
| | - Amanda J Heslegrave
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Tong Guo
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Manuela Tan
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - John Woodside
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Katherine Grosset
- Department of Neurology, Southern General Hospital, University of Glasgow and Institute of Neurological Sciences, Glasgow, UK
| | - Viorica Chelban
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Christine Girges
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Roger A Barker
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - John Hardy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
- Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - Nicholas Wood
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Henry Houlden
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Nigel Williams
- Cardiff University, Cardiff University Institute of Psychological Medicine and Clinical Neurosciences, Cardiff, UK
| | - Yoav Ben-Shlomo
- Department of Social Medicine, University of Bristol, Bristol, UK
| | - Henrik Zetterberg
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Hong Kong Center, for Neurodegenerative Diseases, Hong Kong, People's Republic of China
| | - Donald G Grosset
- Department of Neurology, Southern General Hospital, University of Glasgow and Institute of Neurological Sciences, Glasgow, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
37
|
Detection and assessment of alpha-synuclein in Parkinson disease. Neurochem Int 2022; 158:105358. [PMID: 35561817 DOI: 10.1016/j.neuint.2022.105358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/19/2022] [Accepted: 05/01/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE Different studies have reported varying alpha-synuclein values in the cerebrospinal fluid (CSF), serum, and plasma, making determination of the alpha-synuclein cutoff value for Parkinson's disease difficult and rendering identifying the cause of variation essential. METHOD We searched PubMed from inception to June 2021 and identified 76 eligible studies. Included studies reported data on total, phosphorylated, and oligomeric alpha-synuclein in the CSF, serum, or plasma from individuals with Parkinson's disease and healthy controls. The mean or median alpha-synuclein values from the included studies were summarized and categorized through laboratory assays to visualize potential trends. RESULTS The enzyme-linked immunosorbent assay (ELISA) is the most common assay used to determine alpha-synuclein concentrations. Less common assays include Luminex, single molecule arrays, electrochemiluminescence, and immunomagnetic reduction (IMR). IMR is a single-antibody and wash-free immunoassay designed for determining the extremely low concentration of bio-molecules. For patients with Parkinson's disease, the median or mean testing values ranged from 60.9 to 55,000 pg/mL in the CSF, 0.446 to 1,777,100 pg/mL in plasma, and 0.0292 to 38,200,000 pg/mL in serum. The antibody selection was diverse between studies. The tendency of distribution was more centralized among studies that used the same kit. Studies adopting specific antibodies or in-house assays contribute to the extreme values. Only a few studies on phosphorylated and oligomeric alpha-synuclein were included. CONCLUSION The type of assay and antibody selection in the laboratory played major roles in the alpha-synuclein variation. Studies that used the same assay and kit yielded relatively unanimous results. Furthermore, IMR may be a promising assay for plasma and serum alpha-synuclein quantification. A consensus on sample preparation and testing protocol unification is warranted in the future.
Collapse
|
38
|
Robertson EL, Boehnke SE, Lyra e Silva NDM, Armitage‐Brown B, Winterborn A, Cook DJ, De Felice FG, Munoz DP. Characterization of cerebrospinal fluid biomarkers associated with neurodegenerative diseases in healthy cynomolgus and rhesus macaque monkeys. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12289. [PMID: 35415210 PMCID: PMC8984079 DOI: 10.1002/trc2.12289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Monkeys are becoming important translational models of neurodegenerative disease. To facilitate model development, we measured cerebrospinal fluid (CSF) concentrations of key biomarkers in healthy male and female cynomolgus and rhesus macaques. Amyloid beta (Aβ40, Aβ42), tau (total tau [t-tau], phosphorylated tau [pThr181]), and neurofilament light (NfL) concentrations were measured in CSF of 82 laboratory-housed, experimentally naïve cynomolgus (n = 33) and rhesus (n = 49) macaques. Aβ40 and Aβ42 were significantly higher in rhesus, and female rhesus were higher than males. NfL and t-tau were higher in males, and NfL was higher in rhesus macaques. p-tau was not affected by species or sex. We also examined whether sample location (lumbar or cisterna puncture) affected concentrations. Sample acquisition site only affected NfL, which was higher in CSF from lumbar puncture compared to cisterna magna puncture. Establishing normative biomarker values for laboratory-housed macaque monkeys provides an important resource by which to compare to monkey models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Emma L. Robertson
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
| | - Susan E. Boehnke
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Natalia de M. Lyra e Silva
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Brittney Armitage‐Brown
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Animal Care ServicesQueen's UniversityKingstonOntarioCanada
| | | | - Douglas J. Cook
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of SurgeryKingston Health Sciences CentreKingstonOntarioCanada
| | - Fernanda G. De Felice
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de Janeiro, Cidade Universitaria – Rio de JaneiroRio de JaneiroBrazil
- D'OR Institute for Research and EducationRio de JaneiroBrazil
- Department of PsychiatryProvidence Care HospitalKingstonOntarioCanada
| | - Douglas P. Munoz
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
39
|
Ren J, Pan C, Wang Y, Xue C, Lin H, Xu J, Wang H, Zhang W, Xu P, Chen Y, Liu W. Plasma α-synuclein and phosphorylated tau 181 as a diagnostic biomarker panel for de novo Parkinson's Disease. J Neurochem 2022; 161:506-515. [PMID: 35234288 PMCID: PMC9314946 DOI: 10.1111/jnc.15601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022]
Abstract
The use of a diagnostic panel comprising multiple biomarkers has the potential to accurately diagnose Parkinson’s disease (PD). However, a panel consisting solely of plasma biomarkers to diagnose PD is not available. This study aimed to examine the diagnostic ability of plasma biomarker panels for de novo PD using novel digital ultrasensitive immunoassay technology. We recruited 45 patients with de novo PD and 20 healthy controls (HCs). The concentrations of plasma α‐synuclein (α‐syn), amyloid β‐42 (Aβ42), Aβ40, phosphorylated tau 181 (p‐tau181), neurofilament light (NFL), and glial fibrillary acidic protein (GFAP) were quantified using the ultrasensitive single molecule array (Simoa) platform. Patients with de novo PD had higher plasma levels of α‐syn and p‐tau181 than HCs, adjusting for age and sex. Plasma levels of α‐syn and p‐tau181 were positively correlated in de novo PD patients. Higher plasma α‐syn levels were significantly associated with worse Unified Parkinson’s Disease Rating Scale (UPDRS) Part III motor scores, modified Hoehn and Yahr (H‐Y) stages, and increased risk of PD with mild cognitive impairment (PD‐MCI). Higher plasma p‐tau181 concentrations were linked to worse H‐Y stages. The diagnostic panel using plasma α‐syn and p‐tau181, combined with age and sex, showed good performance in discriminating de novo PD patients from HCs (area under the curve = 0.806). These findings suggest that plasma α‐syn and p‐tau181 together may be a promising diagnostic biomarker panel for de novo PD patients.
Collapse
Affiliation(s)
- Jingru Ren
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chenxi Pan
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yajie Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Xue
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Huixia Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jianxia Xu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Pingyi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong Chen
- Department of Laboratory, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Kwon EH, Tennagels S, Gold R, Gerwert K, Beyer L, Tönges L. Update on CSF Biomarkers in Parkinson's Disease. Biomolecules 2022; 12:biom12020329. [PMID: 35204829 PMCID: PMC8869235 DOI: 10.3390/biom12020329] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
Progress in developing disease-modifying therapies in Parkinson’s disease (PD) can only be achieved through reliable objective markers that help to identify subjects at risk. This includes an early and accurate diagnosis as well as continuous monitoring of disease progression and therapy response. Although PD diagnosis still relies mainly on clinical features, encouragingly, advances in biomarker discovery have been made. The cerebrospinal fluid (CSF) is a biofluid of particular interest to study biomarkers since it is closest to the brain structures and therefore could serve as an ideal source to reflect ongoing pathologic processes. According to the key pathophysiological mechanisms, the CSF status of α-synuclein species, markers of amyloid and tau pathology, neurofilament light chain, lysosomal enzymes and markers of neuroinflammation provide promising preliminary results as candidate biomarkers. Untargeted approaches in the field of metabolomics provide insights into novel and interconnected biological pathways. Markers based on genetic forms of PD can contribute to identifying subgroups suitable for gene-targeted treatment strategies that might also be transferable to sporadic PD. Further validation analyses in large PD cohort studies will identify the CSF biomarker or biomarker combinations with the best value for clinical and research purposes.
Collapse
Affiliation(s)
- Eun Hae Kwon
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
| | - Sabrina Tennagels
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
| | - Klaus Gerwert
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, D-44801 Bochum, Germany
| | - Léon Beyer
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, D-44801 Bochum, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany; (E.H.K.); (S.T.); (R.G.)
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, D-44801 Bochum, Germany; (K.G.); (L.B.)
- Correspondence: ; Tel.: +49-234-509-2420; Fax: +49-234-509-2439
| |
Collapse
|
41
|
Li Q, Li Z, Han X, Shen X, Wang F, Bai L, Li Z, Zhang R, Wang Y, Zhu X. A Panel of Plasma Biomarkers for Differential Diagnosis of Parkinsonian Syndromes. Front Neurosci 2022; 16:805953. [PMID: 35250451 PMCID: PMC8891509 DOI: 10.3389/fnins.2022.805953] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The aim of our study is to explore the most reliable panel of plasma biomarkers for differential diagnosis of parkinsonian syndromes (PDSs). We selected five kinds of neurodegenerative proteins in plasma: neurofilament light chain (NfL), α-synuclein (α-syn), total tau, β-amyloid 42 (Aβ42) and β-amyloid 40 (Aβ40), and investigated the diagnostic value of these biomarkers. Methods A total of 99 plasma samples from patients with Parkinson’s disease (PD), multiple system atrophy (MSA), progressive supranuclear palsy, and age-matched healthy controls (HCs) were enrolled in our study. Plasma NfL, α-syn, total tau, Aβ42, and Aβ40 levels were quantified by ultrasensitive single molecule array immunoassay. We used logistic regression analyses to examine diagnostic accuracy of these plasma biomarkers. Disease severity was assessed by the modified Hoehn and Yahr staging scale, Unified Parkinson’s Disease Rating Scale part III (UPDRS III), and the Mini-Mental State Examination (MMSE), and subsequently, correlation analysis was performed. Results A combination of α-syn, Aβ42, Aβ40, Aβ42/40, and NfL could achieve a best diagnostic value in differentiating PDSs from HC and PD from HC, with an AUC of 0.983 and 0.977, respectively. By adding NfL to measurements of α-syn or Aβ42 or Aβ40 or Aβ42/40, the best discriminating panel was formed in differentiating atypical parkinsonian disorder (APD) and HC, and the discriminatory potential could reach a sensitivity of 100% and specificity of 100% (AUC = 1.000). For further distinguishing PD from APD, we found a combination of NfL, Aβ42, and total tau was the most reliable panel with equally high diagnostic accuracy. With respect to differentiating the subtypes of APD from one another, our results revealed that measurement of NfL, total tau, Aβ42, Aβ40, and Aβ42/40 was the best discriminating panel. Correlation analysis suggests that plasma Aβ42 levels were positively correlated to UPDRS part III scores in MSA. In terms of cognitive function, there was a relationship between plasma Aβ42/40 level and MMSE scores in patients with APD. Conclusion In our study, various combinations of plasma biomarkers have great potentialities in identifying PDSs, with important clinical utility in improving diagnostic accuracy. Plasma NfL may have added value to a blood-based biomarker panel for differentiating PDSs.
Collapse
|
42
|
Murueta-Goyena A, Cipriani R, Carmona-Abellán M, Acera M, Ayo N, del Pino R, Tijero B, Fernández T, Gabilondo I, Zallo F, Matute C, Sánchez-Pernaute R, Khurana V, Cavaliere F, Capetillo-Zarate E, Gómez-Esteban JC. Characterization of molecular biomarkers in cerebrospinal fluid and serum of E46K-SNCA mutation carriers. Parkinsonism Relat Disord 2022; 96:29-35. [DOI: 10.1016/j.parkreldis.2022.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
|
43
|
Huang Y, Huang C, Zhang Q, Shen T, Sun J. Serum NFL discriminates Parkinson disease from essential tremor and reflect motor and cognition severity. BMC Neurol 2022; 22:39. [PMID: 35086487 PMCID: PMC8793176 DOI: 10.1186/s12883-022-02558-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To investigate the diagnostic value of serum neurofilament light chain (NFL) for discriminating Parkinson disease (PD) from Essential tremor (ET) and healthy controls, and to evaluate its correlation with some clinical features of PD patients. METHODS This cross-sectional study measured NFL levels with electrochemiluminescence immunoassay in serum of 146 PD patients, 82 ET patients and 60 age-matched healthy controls. We used multivariate regression analyses to examine whether NFL contributes to PD biomarkers. Disease severity were assessed by Unified Parkinson's Disease Rating Scale part III (UPDRS III), Hoehn & Yahr (H-Y) stage and Mini-Mental State Examination (MMSE). RESULTS Serum NFL levels were significantly higher in PD than in ET and healthy controls (16.6 ± 3.5, 12.2 ± 2.4 and 11.8 ± 2.4 pg/mL, respectively, p < 0.001). In patients with PD, serum NFL were markedly increased in patients with advanced H-Y stage and patients with dementia (both p < 0.001). The correlation analysis revealed that serum NFL was positively associated with UPDRS III score (r = 0.79, p < 0.001) and H-Y stage (r = 0.86, p < 0.001), and negatively correlated with MMSE scores (r = - 0.70, p < 0.001). Further multivariate regression analyses showed that serum NFL was an independent contributor to motor symptom and cognition severity in PD patients (all p < 0.01). CONCLUSIONS Serum NFL levels were markedly elevated may be a useful clinical biomarker for discriminating PD patients from ET and controls. Serum NFL may serve as a potential blood biomarker for motor and cognition severity of PD.
Collapse
Affiliation(s)
- Yixian Huang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, NO. 1055 Sanxiang Road, Suzhou, 215004, China. .,Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Caili Huang
- Department of hematology, The Second Affiliated Hospital of Soochow University, NO. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Qilin Zhang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, NO. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Tingting Shen
- Department of Neurology, The Second Affiliated Hospital of Soochow University, NO. 1055 Sanxiang Road, Suzhou, 215004, China
| | - Jiawei Sun
- Department of Neurology, The Second Affiliated Hospital of Soochow University, NO. 1055 Sanxiang Road, Suzhou, 215004, China
| |
Collapse
|
44
|
Serum neurofilament light chain and postural instability/gait difficulty (PIGD) subtypes of Parkinson’s disease in the MARK-PD study. J Neural Transm (Vienna) 2022; 129:295-300. [PMID: 35072765 PMCID: PMC8930951 DOI: 10.1007/s00702-022-02464-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 11/04/2022]
Abstract
The PIGD (postural instability / gait difficulty) subtype of Parkinson´s disease (PD) is associated with faster cognitive and motor decline. So far, there are no quantifiable biomarkers to aid clinical subtyping. Neurofilament light chain (NfL) is a highly specific marker of neuro-axonal damage and can be assessed in blood. Here, we investigated if serum NfL concentrations are associated with PIGD subtype and PIGD scores in PD patients at advanced disease stages. Furthermore, we evaluated if serum NfL is associated with motor and cognitive function assessed with MDS-UPDRS part III and Montreal cognitive assessment (MoCA). Serum NfL levels were analyzed with Single Molecule Assays (Simoa) in blood of 223 PD patients from the bioMARKers in Parkinson’s Disease (MARK-PD) study. Serum NfL concentrations were higher in PIGD patients independent of age, sex and disease duration. In linear regression analysis, serum NfL levels were associated with MoCA, MDS-UPDRS III and PIGD scores in unadjusted models, but remained significant after adjustment only with PIGD scores. In conclusion, increased serum NfL levels were associated with PIGD subtype and PIGD scores in patients with advanced PD.
Collapse
|
45
|
Diekämper E, Brix B, Stöcker W, Vielhaber S, Galazky I, Kreissl MC, Genseke P, Düzel E, Körtvelyessy P. Neurofilament Levels Are Reflecting the Loss of Presynaptic Dopamine Receptors in Movement Disorders. Front Neurosci 2021; 15:690013. [PMID: 34924923 PMCID: PMC8681873 DOI: 10.3389/fnins.2021.690013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 10/07/2021] [Indexed: 12/01/2022] Open
Abstract
Aims: Neurofilament light chain (NfL) and phosphorylated neurofilament heavy chain (pNfH) are biomarkers for neuroaxonal damage. We assessed whether NfL and other biomarker levels in the CSF are correlated to the loss of presynaptic dopamine transporters in neurons as detected with dopamine transporter SPECT (DaTscan). Methods: We retrospectively identified 47 patients (17 Alzheimer's dementia, 10 idiopathic Parkinson's disease, 7 Lewy body dementia, 13 progressive supranuclear palsy or corticobasal degeneration) who received a DaTscan and a lumbar puncture. DaTscan imaging was performed according to current guidelines, and z-scores indicating the decrease in uptake were software based calculated for the nucleus caudatus and putamen. The CSF biomarkers progranulin, total-tau, alpha-synuclein, NfL, and pNfH were correlated with the z-scores. Results: DaTscan results in AD patients did not correlate with any biomarker. Subsuming every movement disorder with nigrostriatal neurodegeneration resulted in a strong correlation between putamen/nucleus caudatus and NfL (nucleus caudatus right p < 0.01, putamen right p < 0.05, left p < 0.05) and between pNfH and putamen (right p < 0.05; left p < 0.042). Subdividing in disease cohorts did not reveal significant correlations. Progranulin, alpha-synuclein, and total-tau did not correlate with DaTscan results. Conclusion: We show a strong correlation of NfL and pNfH with pathological changes in presynaptic dopamine transporter density in the putamen concomitant to nigrostriatal degeneration. This correlation might explain the reported correlation of impaired motor functions in PD and NfL as seen before, despite the pathological heterogeneity of these diseases.
Collapse
Affiliation(s)
- Elena Diekämper
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Britta Brix
- Institute for Experimental Immunology, EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Winfried Stöcker
- Clinical-Immunological Laboratory Prof. Dr. Stöcker, Lübeck, Germany
| | - Stefan Vielhaber
- Department of Neurology, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Imke Galazky
- Department of Neurology, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Michael C. Kreissl
- Department of Nuclear Medicine, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Philipp Genseke
- Department of Nuclear Medicine, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute for Cognitive Neurology and Dementia Research, Magdeburg, Germany
| | - Péter Körtvelyessy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Neuropathology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
46
|
Chen JH, Chan L, Chung CC, Bamodu OA, Hong CT. Blood Neurofilament Light Chain in Parkinson's Disease: Comparability between Parkinson's Progression Markers Initiative (PPMI) and Asian Cohorts. J Clin Med 2021; 10:jcm10215085. [PMID: 34768602 PMCID: PMC8584818 DOI: 10.3390/jcm10215085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Elevated blood neurofilament light chain (NfL), which indicates the loss of neuronal integrity, is increasingly implicated as a diagnostic and outcome-predicting biomarker for neurological diseases. However, its diagnostic implication for Parkinson’s disease (PD) remains unclear, with conflicting data reported by several studies. This may result from the demographic heterogeneity of the studied cohorts. The present study investigated the comparability of blood NfL between a domestic, single-centered PD cohort from Shuang Ho Hospital (SHH) in Taiwan, with the large international, multi-center cohort, Parkinson’s Progression Markers Initiative (PPMI). In the SHH PD cohort, with 61 people with PD (PwP) and 25 healthy non-PD controls, plasma NfL unexpectedly was significantly higher in the control group than PwP (14.42 ± 13.84 vs. 9.39 ± 6.91 pg/mL, p = 0.05). Interestingly, subgroup analysis revealed a non-significant difference of plasma NfL levels in male PwP compared with controls (8.58 ± 6.21 vs. 7.25 ± 4.43 pg/mL, p =0.575), whereas NfL levels were significantly lower in the female PwP group than in their healthy control peers (10.29 ± 7.62 vs. 17.79 ± 15.52 pg/mL, p = 0.033). Comparative analysis of the SHH and PPMI cohorts revealed a comparable gender-stratified distribution of blood NfL based on approximate theoretical quantiles. After adjusting for age and gender, no apparent difference in NfL value distribution was observed between the SHH and PPMI cohorts’ control or PD groups. Significant downregulation of blood NfL levels were positively correlated with a reduced probability of having a PD diagnosis in both cohorts. These results demonstrated that the adjustment for demographic background enhances comparability between cohorts, and may be required to eliminate covariate/confounder-associated conflict in blood NfL results between different PD studies. This experience may be beneficial to other researchers around the world who are saddled with limited study participants, especially as data from small cohort sizes are often at greater risk of being skewed by specific variables.
Collapse
Affiliation(s)
- Jia-Hung Chen
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (J.-H.C.); (L.C.); (C.-C.C.)
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (J.-H.C.); (L.C.); (C.-C.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (J.-H.C.); (L.C.); (C.-C.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Oluwaseun Adebayo Bamodu
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Department of Hematology and Oncology, Cancer Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Correspondence: (O.A.B.); (C.-T.H.)
| | - Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (J.-H.C.); (L.C.); (C.-C.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Correspondence: (O.A.B.); (C.-T.H.)
| |
Collapse
|
47
|
Zhang L, Gu LY, Dai SB, Zheng R, Jin CY, Fang Y, Yang WY, Tian J, Yin XZ, Zhao GH, Pu JL, Zhang BR, Yan YP, Chen Y. Associations of Body Mass Index-Metabolic Phenotypes with Cognitive Decline in Parkinson's Disease. Eur Neurol 2021; 85:24-30. [PMID: 34689144 DOI: 10.1159/000517538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/19/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Growing evidence suggests important effects of body mass index (BMI) and metabolic status on neurodegenerative diseases. However, the roles of BMI and metabolic status on cognitive outcomes in Parkinson's disease (PD) may vary and are yet to be determined. METHODS In total, 139 PD patients from the whole PD cohort in Parkinson's Progression Markers Initiative database underwent complete laboratory measurements, demographic and anthropometric parameters at baseline, and were enrolled in this study. Further, they were categorized into 4 different BMI-metabolic status phenotypes using Adult Treatment Panel-III criteria. Motor and cognition scales at baseline and longitudinal changes after a 48-month follow-up were compared among the 4 groups. Repeated-measure linear mixed models were performed to compare PD-related biomarkers among BMI-metabolic status phenotypes across time. RESULTS We found that PD patients in the metabolically unhealthy normal weight group showed more cognitive decline in global cognition and visuospatial perception after a 48-month follow-up than those in the other 3 groups (p < 0.05). No difference was found in motor scales among different BMI-metabolic status phenotypes. Finally, compared to the metabolically healthy normal weight group, the metabolically healthy obesity group had lower CSF Aβ42 and serum neurofilament levels in repeated-measure linear mixed models adjusting for age, gender, APOE e4 carrier status, and years of education (p = 0.031 and 0.046, respectively). CONCLUSION The MUNW phenotype was associated with a rapid cognitive decline in PD.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lu-Yan Gu
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shao-Bing Dai
- Department of Anesthesiology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ran Zheng
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chong-Yao Jin
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Fang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wen-Yi Yang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Tian
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xin-Zhen Yin
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Guo-Hua Zhao
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jia-Li Pu
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bao-Rong Zhang
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ya-Ping Yan
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Department of Neurology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
48
|
Niemann L, Lezius S, Maceski A, Leppert D, Englisch C, Schwedhelm E, Zeller T, Gerloff C, Kuhle J, Choe CU. Serum neurofilament is associated with motor function, cognitive decline and subclinical cardiac damage in advanced Parkinson's disease (MARK-PD). Parkinsonism Relat Disord 2021; 90:44-48. [PMID: 34352610 DOI: 10.1016/j.parkreldis.2021.07.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/14/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Serum neurofilament light chain (NfL) levels are associated with disease severity in early Parkinson's disease (PD). We assessed the association of serum NfL with motor and cognitive function and decline in advanced PD patients. METHODS NfL concentrations were analyzed with single molecule array (Simoa) assay in serum of 289 PD patients with advanced disease from the single-center prospective observational biobank study Biomarkers in Parkinson's disease (MARK-PD). Motor and cognitive symptoms were assessed with MDS-UPDRS III, Hoehn&Yahr stages and Montreal Cognitive Assessment (MoCA) at baseline and during 520 [364, 674] days of follow-up. RESULTS Serum NfL concentrations were associated with Hoehn&Yahr stages. During follow-up, baseline NfL levels were associated with time to cognitive decline in adjusted Cox regression models (hazard ratio: 3.23; 95% CI [1.16, 9.00], P < 0.025). Serum NfL was associated with NT-proBNP in adjusted models linking neuronal and cardiac damage in advanced PD patients. CONCLUSION In advanced PD patients, serum NfL concentrations are associated with motor function, cognitive decline and subclinical cardiac damage.
Collapse
Affiliation(s)
- Louisa Niemann
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Lezius
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Aleksandra Maceski
- Department of Biochemistry, University Hospital Basel, Basel, Switzerland
| | - David Leppert
- Department of Biochemistry, University Hospital Basel, Basel, Switzerland
| | - Catrin Englisch
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/ Luebeck, Hamburg, Germany
| | - Tanja Zeller
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/ Luebeck, Hamburg, Germany; Clinic of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Chi-Un Choe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
49
|
Baek MS, Lee MJ, Kim HK, Lyoo CH. Temporal trajectory of biofluid markers in Parkinson's disease. Sci Rep 2021; 11:14820. [PMID: 34285331 PMCID: PMC8292456 DOI: 10.1038/s41598-021-94345-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
Full dynamics of biofluid biomarkers have been unknown in patients with Parkinson’s disease (PD). Using data from 396 PD patients and 182 controls in the Parkinson's Progression Markers Initiative (PPMI) database, we estimated long-term temporal trajectories of CSF α-synuclein (α-syn), amyloid-β (Aβ), total tau (t-tau), phosphorylated tau (p-tau) and serum neurofilament light chain (NfL) by integrating function between the baseline levels and annual changes. At baseline, PD patients showed lower CSF α-syn, Aβ, t-tau and p-tau levels than those of the controls. In all PD patients, CSF α-syn and Aβ decreased in a negative exponential pattern before the onset of motor symptoms, whereas CSF t-tau and p-tau, and serum NfL increased. Patients with cognitive impairment exhibited faster decline of Aβ and α-syn and faster rise of t-tau, p-tau and NfL, when compared to those without. Similarly, low Aβ group showed earlier decline of α-syn, faster rise of t-tau, p-tau and NfL, and faster decline of cognitive performances, when compared to high Aβ group. Our results suggest that longitudinal changes in biomarkers can be influenced by cognitive impairment and Aβ burden at baseline. PD patients with Aβ pathology may be associated with early appearance of α-synuclein pathology, rapid progression of axonal degeneration and neurodegeneration, and consequently greater cognitive decline.
Collapse
Affiliation(s)
- Min Seok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon do, Republic of Korea.,Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Gudeok-ro 179, Seo-gu, Busan, 49241, Republic of Korea.
| | - Han-Kyeol Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, Republic of Korea
| |
Collapse
|
50
|
Abstract
Parkinson's disease is a recognisable clinical syndrome with a range of causes and clinical presentations. Parkinson's disease represents a fast-growing neurodegenerative condition; the rising prevalence worldwide resembles the many characteristics typically observed during a pandemic, except for an infectious cause. In most populations, 3-5% of Parkinson's disease is explained by genetic causes linked to known Parkinson's disease genes, thus representing monogenic Parkinson's disease, whereas 90 genetic risk variants collectively explain 16-36% of the heritable risk of non-monogenic Parkinson's disease. Additional causal associations include having a relative with Parkinson's disease or tremor, constipation, and being a non-smoker, each at least doubling the risk of Parkinson's disease. The diagnosis is clinically based; ancillary testing is reserved for people with an atypical presentation. Current criteria define Parkinson's disease as the presence of bradykinesia combined with either rest tremor, rigidity, or both. However, the clinical presentation is multifaceted and includes many non-motor symptoms. Prognostic counselling is guided by awareness of disease subtypes. Clinically manifest Parkinson's disease is preceded by a potentially long prodromal period. Presently, establishment of prodromal symptoms has no clinical implications other than symptom suppression, although recognition of prodromal parkinsonism will probably have consequences when disease-modifying treatments become available. Treatment goals vary from person to person, emphasising the need for personalised management. There is no reason to postpone symptomatic treatment in people developing disability due to Parkinson's disease. Levodopa is the most common medication used as first-line therapy. Optimal management should start at diagnosis and requires a multidisciplinary team approach, including a growing repertoire of non-pharmacological interventions. At present, no therapy can slow down or arrest the progression of Parkinson's disease, but informed by new insights in genetic causes and mechanisms of neuronal death, several promising strategies are being tested for disease-modifying potential. With the perspective of people with Parkinson's disease as a so-called red thread throughout this Seminar, we will show how personalised management of Parkinson's disease can be optimised.
Collapse
Affiliation(s)
- Bastiaan R Bloem
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Centre of Expertise for Parkinson and Movement Disorders, Nijmegen, Netherlands.
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Christine Klein
- Institute of Neurogenetics and Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|