1
|
Boura I, Poplawska-Domaszewicz K, Limbachiya N, Trivedi D, Batzu L, Chaudhuri KR. Prodromal Parkinson's Disease: A Snapshot of the Landscape. Neurol Clin 2025; 43:209-228. [PMID: 40185519 DOI: 10.1016/j.ncl.2024.12.004] [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
Early observations of specific nonmotor and subtle motor symptoms preceding clinical diagnosis of Parkinson's disease (PD) have paved the way for prodromal PD research, significantly propelling our understanding of early, subclinical stages of neurodegeneration. Prodromal PD has emerged as a complex concept with some researchers suggesting that the period before PD onset is divided into the "at-risk," "preclinical," and "prodromal" phases. Advances in genetic, imaging, laboratory, and digital technologies have enabled the identification of pathophysiological patterns and the potential development of diagnostic, progressive, and therapeutic biomarkers, which could lead to early PD detection and intervention.
Collapse
Affiliation(s)
- Iro Boura
- School of Medicine, University of Crete, Heraklion, Greece; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK.
| | - Karolina Poplawska-Domaszewicz
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Naomi Limbachiya
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Dhaval Trivedi
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Lucia Batzu
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Kallol Ray Chaudhuri
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| |
Collapse
|
2
|
Lan M, Gao M. Association of serum neurofilament light chain and bone mineral density in adults. BMC Musculoskelet Disord 2025; 26:391. [PMID: 40259260 PMCID: PMC12010620 DOI: 10.1186/s12891-025-08639-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Serum neurofilament light chain (sNFL) is a blood-based marker of neuroaxonal damage increasingly used in neurological research. Although sNFL has been linked to systemic aging and chronic disease, its relationship with bone mineral density (BMD) remains unclear. METHODS We analyzed data from 1,344 participants aged ≥ 20 years in the 2013-2014 National Health and Nutrition Examination Survey (NHANES). Serum sNFL concentrations were measured using a high-sensitivity immunoassay. Lumbar BMD was assessed by dual-energy X-ray absorptiometry. Multivariable linear regression models were used to evaluate associations between log-transformed sNFL and BMD, adjusting for demographic, lifestyle, metabolic, renal, cognitive, and bone-related covariates. Sensitivity analyses examined osteoporosis, defined as physician diagnosis or T-score ≤ - 2.5, as a binary outcome. RESULTS Higher sNFL levels were significantly associated with lower lumbar BMD (fully adjusted β = - 0.02 g/cm² per 1-unit increase in ln-sNFL; 95% CI: - 0.04, - 0.01; P = 0.0089). Compared with the lowest quartile, participants in the highest quartile had a 0.04 g/cm² lower BMD (P for trend = 0.011). Sensitivity analyses confirmed higher odds of osteoporosis with increasing sNFL levels (Q4 vs. Q1 OR = 2.70, 95% CI: 1.69, 4.31, P < 0.001). CONCLUSION Elevated serum sNFL concentrations are independently associated with lower lumbar spine BMD in U.S. adults. These findings suggest that sNFL may serve as an exploratory marker of systemic vulnerability relevant to bone health, warranting further longitudinal and mechanistic investigation.
Collapse
Affiliation(s)
- Meihong Lan
- Medical Imaging Center, Shandong Public Health Clinical Center, No.2999, Gangxing West Road, Gaoxin District, Jinan, 250000, Shandong Province, People's Republic of China
| | - Mingming Gao
- Medical Imaging Center, Shandong Public Health Clinical Center, No.2999, Gangxing West Road, Gaoxin District, Jinan, 250000, Shandong Province, People's Republic of China.
| |
Collapse
|
3
|
Liao Y, Zhou K, Lin B, Deng S, Qin L, Weng B, Yang H, Pan L. Associations between blood selenium and serum neurofilament light chain: results of a nationwide survey. Front Neurol 2025; 16:1490760. [PMID: 40264648 PMCID: PMC12011758 DOI: 10.3389/fneur.2025.1490760] [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: 09/09/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Background Selenium (Se) is essential for many nervous system functions including memory, cognition and coordination, which has also been linked to a variety of neurological disorders, such as epilepsy, Alzheimer's disease (AD) and Parkinson's disease (PD). Serum neurofilament light chain (sNfL) is a biomarker of neurologic diseases. Studies on the relationship between blood Se and sNfL are limited. Methods The National Health and Nutrition Examination Survey (NHANES) 2013-2014 data were employed to perform multivariate linear regression analysis and smooth curve fitting in order to investigate the relationship between blood Se and sNfL. Utilizing subgroup analyses and interaction tests, the stability of this relationship between populations was evaluated. Results sNfL and blood Se had an inverse relationship in 1,036 individuals who were older than 20. According to the fully adjusted model, the sNfL decreased by 54.75 pg./mL for every unit increase in log blood Se [β = -54.75, 95% CI (-75.36, -34.14)]. The sNfL of individuals in the highest blood Se quartile decreased by 3.4 pg./mL in comparison to those in the lowest quartile [β = -3.40, 95% CI (-6.47, -0.32)]. This inverse association was more significant in those who were younger than 60 years old, male, normal weight, had a history of smoking and drinking. Conclusion Blood Se is inversely associated with sNfL in American adults. Our findings indicate that blood Se may have a potential protective effect against neuronal damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Liya Pan
- Department of Neurology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
4
|
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
|
5
|
Dilixiati D, Kadier K, Qiao B, Zhang W, Nuerdebieke D, Zebibula A, Yang Y, Rexiati M. Association between serum neurofilament light chain levels and grip strength among US adults: a cross-sectional study using National Health and Nutrition Examination Survey data from 2013 to 2014. BMJ Open 2025; 15:e084766. [PMID: 40180396 PMCID: PMC11969582 DOI: 10.1136/bmjopen-2024-084766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 01/15/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVE We aimed to investigate the relationship between serum neurofilament light chain (NfL) and grip strength using data from the 2013-2014 US National Health and Nutrition Examination Survey (NHANES). DESIGN Secondary analysis of cross-sectional, population-based data. SETTING NHANES sample, 2013-2014. PARTICIPANTS We studied 1925 participants aged 20-75 years. OUTCOME MEASURES AND ANALYSIS We applied a multivariable generalised linear regression model, adjusted for several potential confounders, and restrictive cubic spline models to evaluate the association between serum NfL and grip strength. Subgroup analyses were conducted using stratified multivariable linear regression analysis. RESULTS We included 1925 participants (average age: 44.8±0.44 years) from the NHANES database. Participants with higher serum NfL levels had a significantly higher prevalence of medical conditions (hypertension, diabetes, cardiovascular disorder, chronic kidney disease (CKD) and cancer) compared with those with lower NfL levels (all p<0.001). After adjusting for confounding factors, there was a negative association between serum NfL and grip strength (β=-2.07; 95% CI -3.47, -0.67; p=0.007). In addition, significant interactions were found between NfL and grip strength stratified by age, physical activity and CKD (p value for interaction=0.002, 0.023 and 0.006). The results of the restricted cubic splines (RCS) analysis showed no evidence against a linear association of serum NfL levels with grip strength. (p for non-linearity=0.334). CONCLUSION Our study demonstrates a strong, negative and linear correlation between elevated serum NfL levels and grip strength. Notably, our findings indicate that individuals aged between 60 and 75 years, those with physical inactivity and those with CKD exhibit a more pronounced reduction in grip strength with increasing serum NfL levels.
Collapse
Affiliation(s)
- Diliyaer Dilixiati
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Kaisaierjiang Kadier
- Department of Cardiology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Bingzhang Qiao
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Weijie Zhang
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Daniyaer Nuerdebieke
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Abudureheman Zebibula
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Ying Yang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mulati Rexiati
- Department of Urology, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| |
Collapse
|
6
|
Rissardo JP, Caprara ALF. A Narrative Review on Biochemical Markers and Emerging Treatments in Prodromal Synucleinopathies. Clin Pract 2025; 15:65. [PMID: 40136601 PMCID: PMC11941140 DOI: 10.3390/clinpract15030065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Alpha-synuclein has been associated with neurodegeneration, especially in Parkinson's disease (PD). This study aimed to review clinical, biochemical, and neuroimaging markers and management of prodromal synucleinopathies. The prodromal state of synucleinopathies can be better understood with PD pathophysiology, and it can be separated into premotor and pre-diagnostic phases. The incidence of PD in patients with prodromal phase symptoms ranges from 0.07 to 14.30, and the most frequently studied pathology is the REM behavioral disorder (RBD). Neuroimaging markers are related to dopamine denervation, brain perfusion changes, gross anatomy changes, and peripheral abnormalities. α-synuclein assays (SAA) in CSF revealed high sensitivity (up to 97%) and high specificity (up to 92%); in the last decade, there was the development of other matrices (blood, skin, and olfactory mucosa) for obtaining quantitative and qualitative α-synuclein. Other biomarkers are neurofilament light chain, DOPA decarboxylase, and multiplexed mass spectrometry assay. Regarding genetic counseling in α-synucleinopathies, it is an important topic in clinical practice to discuss with patients with high-risk individuals and should involve basic principles of autonomy, beneficence, and non-maleficence. Some of the themes that should be reviewed are the involvement of physical activity, diet (including alcohol, coffee, and vitamin supplementation), smoking, sleep, and stress in the pathophysiology of synucleinopathies. The number of trials related to prodromal symptoms is still scarce, and the number of studies evaluating intervention is even lower.
Collapse
|
7
|
Oizumi H, Hasegawa T, Kawahata I, Sekimori T, Totsune T, Sugimura Y, Baba T, Fukunaga K, Takeda A. Associations among blood biomarkers, clinical subtypes, and prognosis in Parkinson's disease. Clin Park Relat Disord 2025; 12:100313. [PMID: 40161513 PMCID: PMC11952764 DOI: 10.1016/j.prdoa.2025.100313] [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: 10/14/2024] [Revised: 01/30/2025] [Accepted: 03/02/2025] [Indexed: 04/02/2025] Open
Abstract
Background Early identification of the poor prognosis subtype by surrogate markers would be advantageous for selecting treatments for Parkinson's disease (PD). The aim of the present study was to test whether plasma neurofilament light chain (NF-L), total tau (t-tau), ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1), fatty acid-binding protein 3 (FABP3), and phosphorylated tau (p-tau) can be used as prognostic biomarkers in PD. Methods In the present study, both retrospective and prospective studies were performed. Plasma samples at baseline from 81 PD patients were included in the prospective study. Plasma samples at baseline from 60 patients who underwent cognitive assessment were subjected to the hierarchical cluster analysis for a retrospective study. Results On the basis of the results of the cluster analysis, patients were classified into three groups: groups (G)1, G2 and G3. Individuals in the G1 cluster, who had an older age at onset and were prone to early progression with dementia, had significantly greater plasma NF-L levels than those in the G3 cluster, who did not present with dementia at an early stage. A Cox proportional hazards regression model adjusted for age and sex revealed that high NF-L and UCH-L1 levels at baseline predicted the four future milestones (i.e., nursing care, dysphagia, wheelchair use, and repeated falls), and high plasma t-tau at baseline predicted future dysphagia. Conclusions Although further studies with a larger number of patients will be required, plasma NF-L may be a useful biomarker for identifying the rapidly progressive subtype of PD, and plasma NF-L and UCH-L1 may serve as biomarkers of overall PD prognosis, whereas plasma t-tau could be a biomarker for future dysphagia in PD.
Collapse
Affiliation(s)
- Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
| | - Takafumi Hasegawa
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
- Department of Neurology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Tohoku University Graduate School of Pharmaceutical Sciences, 6-3, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University Graduate School of Medicine, Hikarigaoka, Fukushima City, Fukushima 960-1295, Japan
| | - Tomoki Sekimori
- Department of CNS Drug Innovation, Tohoku University Graduate School of Pharmaceutical Sciences, 6-3, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tomoko Totsune
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
| | - Yoko Sugimura
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
| | - Toru Baba
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Tohoku University Graduate School of Pharmaceutical Sciences, 6-3, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, 2-11-11, Kagitorihoncho, Taihaku-ku, Sendai, Miyagi 982-8555, Japan
- Department of Cognitive and Motor Aging, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
8
|
Chahine LM, Lafontant DE, Choi SH, Iwaki H, Blauwendraat C, Singleton AB, Brumm MC, Alcalay RN, Merchant K, Nudelman KNH, Dagher A, Vo A, Tao Q, Venuto CS, Kieburtz K, Poston KL, Bressman S, Gonzalez-Latapi P, Avants B, Coffey C, Jennings D, Tolosa E, Siderowf A, Marek K, Simuni T. LRRK2-associated parkinsonism with and without in vivo evidence of alpha-synuclein aggregates: longitudinal clinical and biomarker characterization. Brain Commun 2025; 7:fcaf103. [PMID: 40114783 PMCID: PMC11925012 DOI: 10.1093/braincomms/fcaf103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Among LRRK2-associated parkinsonism cases with nigral degeneration, over two-thirds demonstrate evidence of pathologic alpha-synuclein, but many do not. Understanding the clinical phenotype and underlying biology in such individuals is critical for therapeutic development. Our objective was to compare clinical and biomarker features, and rate of progression over 4 years of follow-up, among LRRK2-associated parkinsonism cases with and without in vivo evidence of alpha-synuclein aggregates. Data were from the Parkinson's Progression Markers Initiative, a multicentre prospective cohort study. The sample included individuals diagnosed with Parkinson disease with pathogenic variants in LRRK2. Presence of CSF alpha-synuclein aggregation was assessed with seed amplification assay. A range of clinician- and patient-reported outcome assessments were administered. Biomarkers included dopamine transporter scan, CSF amyloid-beta1-42, total tau, phospho-tau181, urine bis(monoacylglycerol)phosphate levels and serum neurofilament light chain. Linear mixed-effects (LMMs) models examined differences in trajectory in CSF-negative and CSF-positive groups. A total of 148 LRRK2 parkinsonism cases (86% with G2019S variant), 46 negative and 102 positive for CSF alpha-synuclein seed amplification assay, were included. At baseline, the negative group was older than the positive group [median (inter-quartile range) 69.1 (65.2-72.3) versus 61.5 (55.6-66.9) years, P < 0.001] and a greater proportion were female [28 (61%) versus 43 (42%), P = 0.035]. Despite being older, the negative group had similar duration since diagnosis and similar motor rating scale [16 (11-23) versus 16 (10-22), P = 0.480] though lower levodopa equivalents. Only 13 (29%) of the negative group were hyposmic, compared with 75 (77%) of the positive group. The negative group, compared with the positive group, had higher per cent-expected putamenal dopamine transporter binding for their age and sex [0.36 (0.29-0.45) versus 0.26 (0.22-0.37), P < 0.001]. Serum neurofilament light chain was higher in the negative group compared with the positive group [17.10 (13.60-22.10) versus 10.50 (8.43-14.70) pg/mL; age-adjusted P-value = 0.013]. In terms of longitudinal change, the negative group remained stable in functional rating scale score in contrast to the positive group who had a significant increase (worsening) of 0.729 per year (P = 0.037), but no other differences in trajectory were found. Among individuals diagnosed with Parkinson disease with pathogenic variants in the LRRK2 gene, we found clinical and biomarker differences in cases without versus with in vivo evidence of CSF alpha-synuclein aggregates. LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates as a group exhibit less severe motor manifestations and decline. The underlying biology in LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates requires further investigation.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David-Erick Lafontant
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Seung Ho Choi
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Hirotaka Iwaki
- DataTecnica LLC, Washington, DC 20037, USA
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew B Singleton
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Roy N Alcalay
- Tel Aviv Sourasky Medical Center, 64239 Tel-Aviv, Israel
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Andrew Vo
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Qin Tao
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada H3A 2B4
| | - Charles S Venuto
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY 14642,USA
| | - Karl Kieburtz
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY 14642,USA
| | - Kathleen L Poston
- Department of Neurology, Stanford University School of Medicine, Palo Alto, 94304 CA, USA
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine, Mount Sinai Beth Israel, New York City, NY 10029, USA
| | - Paulina Gonzalez-Latapi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 55848, USA
| | - Danna Jennings
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eduardo Tolosa
- Parkinson’s Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, 08028 Barcelona, Spain
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ken Marek
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA
| | - Tatyana Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
9
|
Gao S, Wang Z, Huang Y, Yang G, Wang Y, Yi Y, Zhou Q, Jian X, Zhao G, Li B, Xu L, Xia K, Tang B, Li J. Early detection of Parkinson's disease through multiplex blood and urine biomarkers prior to clinical diagnosis. NPJ Parkinsons Dis 2025; 11:35. [PMID: 39994191 PMCID: PMC11850829 DOI: 10.1038/s41531-025-00888-2] [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: 08/30/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Blood and urine biomarkers are commonly used to diagnose and monitor chronic diseases. We initially screened 67 biomarkers, including 4 urine biomarkers and 63 blood biomarkers, and identified 13 blood biomarkers significantly associated with Parkinson's disease (PD). Among these, we discovered three novel markers demonstrating strong associations: phosphate (P = 1.81 × 10-3), AST/ALT ratio (P = 8.53 × 10-6), and immature reticulocyte fraction (IRF) (P = 3.49 × 10-20). We also substantiated eight well-studied biomarkers and elucidated the roles of two previously ambiguous biomarkers. Our analyses confirmed IGF-1 (P = 7.46 × 10-29) as a risk factor, and C-reactive protein (CRP) (P = 1.43 × 10-3) as protective against PD. Genetic analysis highlighted that IRF, CRP, and IGF-1 share significant genetic loci with PD, notably at MAPT, SETD1A, HLA-DRB1, and HLA-DQA1. Furthermore, Mendelian randomization (MR) analysis suggested potential causal associations between IGF-1, CRP, and PD. We identified several blood biomarkers that may be associated with the risk of developing PD, providing valuable insights for further exploration of PD-related biomarkers.
Collapse
Affiliation(s)
- Shuo Gao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zheng Wang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuanfeng Huang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Guang Yang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yijing Wang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
| | - Yan Yi
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
| | - Qiao Zhou
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xingxing Jian
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Linyong Xu
- Hunan Provincial Key Laboratory of Oral Health Research & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Kun Xia
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Beisha Tang
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
10
|
Lin JB, El Helwe H, Falah H, Hammerschlag BL, Schultz SA, Baldwin G, Xue Y, Vasan RA, Song C, Lo K, Meeker A, Wang SL, Kivisäkk P, Solá-Del Valle D, Margeta MA. Evaluation of Serum and Aqueous Humor Neurofilament Light Chain as Markers of Neurodegeneration in Glaucoma. Transl Vis Sci Technol 2025; 14:24. [PMID: 39998458 PMCID: PMC11875033 DOI: 10.1167/tvst.14.2.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/19/2025] [Indexed: 02/26/2025] Open
Abstract
Purpose The purpose of this study was to evaluate the relationship between serum and aqueous humor (AH) neurofilament light chain (NfL) and to determine whether serum NfL is elevated in patients undergoing ocular surgery who have glaucoma compared with those who do not. Methods In this single-center, case-control study, we enrolled patients with various types and stages of glaucoma undergoing planned ophthalmic surgery as part of their routine care and compared them with patients without glaucoma undergoing phacoemulsification for age-related cataract. We recruited 110 patients with glaucoma and 113 patients without glaucoma and collected AH and blood from these participants. Levels of AH and serum NfL were quantified using the Single-Molecule Array (Simoa) NF-light assay (Quanterix). Clinical information was obtained by reviewing the medical records. Results In a model controlling for age and body mass index (BMI), AH NfL was significantly elevated in patients with glaucoma compared with controls (P < 0.001). In contrast, after controlling for age, BMI, and Mini Mental Status Examination (MMSE) scores, serum NfL was not elevated in patients with glaucoma compared with controls (P = 0.81). Conclusions Although our findings validate AH NfL as a marker of glaucomatous neurodegeneration, no such evidence was found for serum NfL. Translational Relevance NfL levels in AH may be a molecular marker of retinal ganglion cell health in glaucoma; in contrast, serum NfL has limited utility for monitoring glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Jonathan B. Lin
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Hani El Helwe
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Henisk Falah
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | | | | | - George Baldwin
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Yixi Xue
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Ryan A. Vasan
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Christian Song
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Kristine Lo
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Austin Meeker
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Silas L. Wang
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David Solá-Del Valle
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| | - Milica A. Margeta
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA, USA
| |
Collapse
|
11
|
Hong CT, Chung CC, Hsieh YC, Chan L. Plasma extracellular vesicle neurofilament light chain as the biomarkers of the progression of Parkinson's disease. BIOMOLECULES & BIOMEDICINE 2025; 25:588-594. [PMID: 39652080 PMCID: PMC12010974 DOI: 10.17305/bb.2024.11502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 01/31/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by progressive symptoms, underscoring the urgent need for predictive blood biomarkers. Plasma extracellular vesicles (EVs) offer a promising platform for biomarker development, with neurofilament light chain (NfL) emerging as a potential candidate for neurological diseases. This study evaluated plasma EV NfL as a biomarker for disease progression in a PD cohort.A total of 55 patients with PD (PwP) and 58 healthy controls (HCs) were followed, with PwP completing an average of 3.96 visits and HCs 2.25 visits. Plasma EVs were isolated and validated, and EV NfL levels were measured using an immunomagnetic reduction assay. Generalized estimating equations and Spearman correlations assessed relationships between clinical symptom progression and biomarkers. Although no significant differences in plasma EV NfL levels were observed between PwP and HCs over time, changes in plasma EV NfL significantly correlated with motor symptom progression, specifically with adjusted-total and akinetic-rigidity subscores of the Unified PD Rating Scale (UPDRS) Part III. Additionally, changes in UPDRS Part II scores were significantly associated with plasma EV NfL levels. These findings suggest that plasma EV NfL reflects motor symptom progression in PwP, highlighting its potential as a valuable biomarker for monitoring disease progression and guiding clinical trials in PD.
Collapse
Affiliation(s)
- Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chen Hsieh
- College of Medical Science and Technology, Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Marzetti E, Di Lorenzo R, Calvani R, Pesce V, Landi F, Coelho-Júnior HJ, Picca A. From Cell Architecture to Mitochondrial Signaling: Role of Intermediate Filaments in Health, Aging, and Disease. Int J Mol Sci 2025; 26:1100. [PMID: 39940869 PMCID: PMC11817570 DOI: 10.3390/ijms26031100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
The coordination of cytoskeletal proteins shapes cell architectures and functions. Age-related changes in cellular mechanical properties have been linked to decreased cellular and tissue dysfunction. Studies have also found a relationship between mitochondrial function and the cytoskeleton. Cytoskeleton inhibitors impact mitochondrial quality and function, including motility and morphology, membrane potential, and respiration. The regulatory properties of the cytoskeleton on mitochondrial functions are involved in the pathogenesis of several diseases. Disassembly of the axon's cytoskeleton and the release of neurofilament fragments have been documented during neurodegeneration. However, these changes can also be related to mitochondrial impairments, spanning from reduced mitochondrial quality to altered bioenergetics. Herein, we discuss recent research highlighting some of the pathophysiological roles of cytoskeleton disassembly in aging, neurodegeneration, and neuromuscular diseases, with a focus on studies that explored the relationship between intermediate filaments and mitochondrial signaling as relevant contributors to cellular health and disease.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (E.M.); (R.C.); (F.L.); (H.J.C.-J.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Rosa Di Lorenzo
- Department of Biosciences, Biotechnologies and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (E.M.); (R.C.); (F.L.); (H.J.C.-J.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Vito Pesce
- Department of Biosciences, Biotechnologies and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy; (R.D.L.); (V.P.)
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (E.M.); (R.C.); (F.L.); (H.J.C.-J.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Hélio José Coelho-Júnior
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (E.M.); (R.C.); (F.L.); (H.J.C.-J.)
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (E.M.); (R.C.); (F.L.); (H.J.C.-J.)
- Department of Medicine and Surgery, LUM University, Str. Statale 100, 70010 Casamassima, Italy
| |
Collapse
|
13
|
Pehlivan VF, Pehlivan B, Celik H, Duran E, Taskın A, Taskın S, Tatlı F. Investigation of the Acute Effects of Two Different Preoxygenation Methods on Neurodegenerative Biomarkers in Laparoscopic Cholecystectomy Surgery. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:167. [PMID: 40005285 PMCID: PMC11857148 DOI: 10.3390/medicina61020167] [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] [Received: 12/03/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Oxygen is essential for all living organisms and plays a critical role in anesthesia and intensive care practices. However, the notion that unlimited oxygen therapy is harmless is a misconception. Our study investigates the acute effects of different preoxygenation methods on hemodynamic parameters and neurodegenerative biomarkers in patients undergoing laparoscopic cholecystectomy surgery. Materials and Methods: This prospective, randomized, controlled study included 52 patients undergoing elective laparoscopic cholecystectomy under general anesthesia. Patients were divided into two groups: Group I received standard preoxygenation (100% FiO2 for 3 min), while Group II underwent rapid preoxygenation (eight deep breaths over 30 s to 1 min). Hemodynamic parameters (SAP, DAP, MAP, and SpO2) and neurodegenerative biomarkers (pTau, S100B, NSE, NfL, GFAP) were measured after preoxygenation, after intubation, and at the end of surgery. Results: Group I exhibited a significant increase in levels of pTau, S100B, NSE, and GFAP, indicating higher neuronal and glial cell stress compared to Group II (p < 0.001). No significant increase in NfL levels was observed in either group. Hemodynamic parameters (HR, SAP, DAP, MAP) were significantly higher during and after preoxygenation in Group I, suggesting an increased stress response. Group II showed lower levels of acute neurotoxicity and oxidative stress. Conclusions: Our findings indicate that preoxygenation with 100% FiO2 induces stress in neuronal cells, axons, and glial cells, leading to an increase in neurodegenerative biomarkers. Optimizing preoxygenation strategies is crucial to reduce oxidative stress and improve neurological outcomes for surgical patients.
Collapse
Affiliation(s)
- Veli Fahri Pehlivan
- Department of Anesthesia and Reanimation, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; (B.P.); (E.D.)
| | - Basak Pehlivan
- Department of Anesthesia and Reanimation, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; (B.P.); (E.D.)
| | - Hakim Celik
- Department of Physiology, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; (H.C.); (S.T.)
| | - Erdogan Duran
- Department of Anesthesia and Reanimation, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; (B.P.); (E.D.)
| | - Abdullah Taskın
- Department of Nutrition and Dietetics, Health Science Faculty, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey;
| | - Seyhan Taskın
- Department of Physiology, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; (H.C.); (S.T.)
| | - Faik Tatlı
- Department of General Surgery, Faculty of Medicine, Harran University, Osmanbey Campus, PC 63300 Sanliurfa, Turkey; faiktatli-@hotmail.com
| |
Collapse
|
14
|
Alberti G, Portelli D, Polito F, Graceffa A, Licitri L, Loteta S, Torre MM, Gasparo I, Rizzo V, Aguennouz M, Macaione V. Blood Neurofilament Light Chain and Phospho-Tau 181 in Subjects with Mild Cognitive Impairment Due to Age-Related Hearing Loss. J Clin Med 2025; 14:672. [PMID: 39941343 PMCID: PMC11818439 DOI: 10.3390/jcm14030672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Mild cognitive impairment is increasingly recognized as a precursor to more severe neurodegenerative conditions, particularly in the context of aging. Recent studies have highlighted the intersection of hearing loss and cognitive decline, suggesting that auditory deficits may exacerbate cognitive impairments in older adults, proposing the use of hearing aids to mitigate cognitive decline, and indicating that early intervention in hearing loss could be crucial for preserving cognitive function. The underlying mechanisms of the relationship between hearing and cognitive impairment may involve neuroinflammatory processes and neurodegeneration. Recent studies have evidenced the role of tau proteins and neurofilaments as biomarkers in the onset and progression of neurodegenerative diseases. Methods: We selected 30 subjects with age-related hearing loss, and we evaluated their cognitive status through the administration of screening tests, which also measured neurofilament light chain and phospho-tau 181 serum levels as biomarkers of neurodegeneration. The subjects were re-evaluated six months after the hearing aid fitting. Results: Patients with hearing impairment presented slightly altered results on cognitive tests, typical of a mild cognitive impairment. At the same time, serum levels of neurofilament light chain and phospho-tau 181 were significantly increased compared to the matched control group. After the hearing aids fitting, auditory, cognitive, and serum values results improved. Conclusions: The results of the study highlight the cognitive involvement in patients with hearing impairment and identify neurofilament light chain and phospho-tau 181 as serum biomarkers of neurodegeneration useful in monitoring the pathology.
Collapse
Affiliation(s)
- Giuseppe Alberti
- Department of Adult and Development Age Human Pathology, University of Messina, 98122 Messina, Italy; (G.A.); (D.P.); (S.L.)
| | - Daniele Portelli
- Department of Adult and Development Age Human Pathology, University of Messina, 98122 Messina, Italy; (G.A.); (D.P.); (S.L.)
| | - Francesca Polito
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Anita Graceffa
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Laura Licitri
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Sabrina Loteta
- Department of Adult and Development Age Human Pathology, University of Messina, 98122 Messina, Italy; (G.A.); (D.P.); (S.L.)
| | - Margherita Maria Torre
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Irene Gasparo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Vincenzo Rizzo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - M’hammed Aguennouz
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| | - Vincenzo Macaione
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (F.P.); (A.G.); (L.L.); (M.M.T.); (I.G.); (V.R.); (M.A.)
| |
Collapse
|
15
|
Gu L, Zhang P, Gao R, Shu H, Wang P. Predictive value of serum neurofilament light chain for cognitive impairment in Parkinson's disease. Front Aging Neurosci 2024; 16:1465016. [PMID: 39703922 PMCID: PMC11655485 DOI: 10.3389/fnagi.2024.1465016] [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: 07/15/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
Background Neurofilament light chain (NfL) has recently emerged as a key indicator of neurodegeneration. In this study, our hypothesis is that the levels of blood-derived NfL and its accumulation during the Parkinson's disease (PD) progression could serve as a potential biomarker for predicting subsequent cognitive decline. To investigate this, we conducted a study utilizing a large single-center cohort. Methods The study included 193 participants, consisting of 106 cognitively normal PD (PD-CN) patients and 87 normal controls (NC) individuals. Serum NfL concentrations were measured. PD patients were followed up for clinical assessment at an average of 2 ± 0.6 years. Results The serum NfL levels were significantly higher in PD-CN patients compared to NC. PD-CN patients and NC at follow-up time exhibited higher serum NfL levels compared to those at baseline. PD patients with high serum NfL levels were found to have a higher likelihood of transitioning from normal cognition to mild cognitive impairment (MCI) or dementia (Hazard ratio (HR) 1.107, 95% confidence intervals (CI) 1.010-1.213, p = 0.030). The area under the curve (AUC) for PD-CN conversion to MCI or dementia at follow-up time was determined to be 0.684 (95% CI 0.569-0.799). Conclusion In conclusion, our study found that PD patients have significantly higher levels of serum NfL compared to individuals without PD. Furthermore, serum NfL levels increase as PD progresses and can predict cognitive impairment within a 2-year timeframe. Serum NfL may serve as a feasible, non-invasive biomarker of cognitive progression in PD. However, further studies and functional experiments are needed to validate these findings.
Collapse
Affiliation(s)
- Lihua Gu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Pengcheng Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Rui Gao
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Hao Shu
- Department of Neurology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
16
|
Sharma P, Giri A, Tripathi PN. Emerging Trends: Neurofilament Biomarkers in Precision Neurology. Neurochem Res 2024; 49:3208-3225. [PMID: 39347854 DOI: 10.1007/s11064-024-04244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024]
Abstract
Neurofilaments are structural proteins found in the cytoplasm of neurons, particularly in axons, providing structural support and stability to the axon. They consist of multiple subunits, including NF-H, NF-M, and NF-L, which form long filaments along the axon's length. Neurofilaments are crucial for maintaining the shape and integrity of neurons, promoting axonal transport, and regulating neuronal function. They are part of the intermediate filament (IF) family, which has approximately 70 tissue-specific genes. This diversity allows for a customizable cytoplasmic meshwork, adapting to the unique structural demands of different tissues and cell types. Neurofilament proteins show increased levels in both cerebrospinal fluid (CSF) and blood after neuroaxonal damage, indicating injury regardless of the underlying etiology. Precise measurement and long-term monitoring of damage are necessary for determining prognosis, assessing disease activity, tracking therapeutic responses, and creating treatments. These investigations contribute to our understanding of the importance of proper NF composition in fundamental neuronal processes and have implications for neurological disorders associated with NF abnormalities along with its alteration in different animal and human models. Here in this review, we have highlighted various neurological disorders such as Alzheimer's, Parkinson's, Huntington's, Dementia, and paved the way to use neurofilament as a marker in managing neurological disorders.
Collapse
Affiliation(s)
- Priti Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India
| | - Aditi Giri
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
| | - Prabhash Nath Tripathi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
17
|
Devarakonda SS, Basha S, Pithakumar A, L B T, Mukunda DC, Rodrigues J, K A, Biswas S, Pai AR, Belurkar S, Mahato KK. Molecular mechanisms of neurofilament alterations and its application in assessing neurodegenerative disorders. Ageing Res Rev 2024; 102:102566. [PMID: 39481763 DOI: 10.1016/j.arr.2024.102566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Neurofilaments are intermediate filaments present in neurons. These provide structural support and maintain the size and shape of the neurons. Dysregulation, mutation, and aggregation of neurofilaments raise the levels of these proteins in the blood and cerebrospinal fluid (CSF), which are characteristic features of axonal damage and certain rare neurological diseases, such as Giant Axonal Neuropathy and Charcot-Mare-Tooth disease. Understanding the structure, dynamics, and function of neurofilaments has been greatly enhanced by a diverse range of biochemical and preclinical investigations conducted over more than four decades. Recently, there has been a resurgence of interest in post-translational modifications of neurofilaments, such as phosphorylation, aggregation, mutation, oxidation, etc. Over the past twenty years, several rare disorders have been studied from structural alterations of neurofilaments. These disorders are monitored by fluid biomarkers such as neurofilament light chains. Currently, there are many tools, such as Enzyme-Linked Immunosorbent Assay, Electrochemiluminescence Assay, Single-Molecule Array, Western/immunoblotting, etc., in use to assess the neurofilament proteins in Blood and CSF. However, all these techniques utilize expensive, non-specific, or antibody-based methods, which make them unsuitable for routine screening of neurodegenerative disorders. This provides room to search for newer sensitive, cost-effective, point-of-care tools for rapid screening of the disease. For a long time, the molecular mechanisms of neurofilaments have been poorly understood due to insufficient research attempts, and a deeper understanding of them remains elusive. Therefore, this review aims to highlight the available literature on molecular mechanisms of neurofilaments and the function of neurofilaments in axonal transport, axonal conduction, axonal growth, and neurofilament aggregation, respectively. Further, this review discusses the role of neurofilaments as potential biomarkers for the identification of several neurodegenerative diseases in clinical laboratory practice.
Collapse
Affiliation(s)
| | - Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Anjana Pithakumar
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Thoshna L B
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Ameera K
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Shimul Biswas
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Sushma Belurkar
- Department of Pathology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India.
| |
Collapse
|
18
|
Pilotto A, Ashton NJ, Lupini A, Battaglio B, Zatti C, Trasciatti C, Gipponi S, Cottini E, Grossi I, Salvi A, de Petro G, Pizzi M, Canale A, Blennow K, Zetterberg H, Padovani A. Plasma NfL, GFAP, amyloid, and p-tau species as Prognostic biomarkers in Parkinson's disease. J Neurol 2024; 271:7537-7546. [PMID: 39249107 PMCID: PMC11588809 DOI: 10.1007/s00415-024-12669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION The prognostic role of plasma neurofilament light chain (NfL), phospho-tau, beta-amyloid, and GFAP is still debated in Parkinson's disease (PD). METHODS Plasma p-tau181, p-tau231, Aβ1-40, Aβ1-42, GFAP, and NfL were measured by SIMOA in 136 PD with 2.9 + 1.7 years of follow-up and 76 controls. Differences in plasma levels between controls and PD and their correlation with clinical severity and progression rates were evaluated using linear regression analyses. RESULTS Patients exhibited similar distribution of plasma biomarkers but higher P-tau181, P-tau231 and lower Aβ1-42 compared with controls. NfL and GFAP correlated with baseline motor and non-motor severity measures. At follow-up, NfL emerged as the best predictor of progression with marginal effect of GFAP and p-tau181 adjusting for age, sex, disease duration, and baseline motor severity. CONCLUSION The present findings confirmed plasma NfL as best predictor of progression in PD, with a marginal role of p-tau181 and GFAP.
Collapse
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy.
- Department of Continuity of Care and Frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy.
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy.
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Alessandro Lupini
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
| | - Beatrice Battaglio
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
| | - Cinzia Zatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
| | - Chiara Trasciatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
- Department of Continuity of Care and Frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Stefano Gipponi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
| | - Elisabetta Cottini
- Department of Continuity of Care and Frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
| | - Ilaria Grossi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Salvi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giuseppina de Petro
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Canale
- Department of Statistical Sciences, University of Padova, Padua, Italy
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 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, Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, People's Republic of China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, P.Zzale Spedali Civili, 1, 25123, Brescia, Italy
- Department of Continuity of Care and Frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| |
Collapse
|
19
|
Minibajeva O, Karelis G, Zolovs M, Ķēniņa V. Human Leukocyte Antigen Polymorphism and Blood Biomarker Profiles in Parkinson's Disease: A Pilot Study in a Latvian Cohort. Biomedicines 2024; 12:2709. [PMID: 39767615 PMCID: PMC11673695 DOI: 10.3390/biomedicines12122709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Parkinson's disease (PD) is a neurodegenerative disorder characterised by a high prevalence of sporadic cases. Various molecular mechanisms are involved in its pathogenesis. This pilot study aimed to identify potential risk and protective human leukocyte antigen (HLA) alleles in PD, discover candidate alleles for further research, and evaluate potential blood biomarkers. Methods: A total of 43 PD patients and 79 unrelated sex-matched controls were enrolled in this study. We analysed the polymorphism of HLA-DRB1, HLA-DQA1, and HLA-DQB1 alleles and the blood levels of biomarkers such as S100 calcium-binding protein A9 (S1000A9), kynurenic acid (KYNA), neurofilament light chain (NfL), and glutamate decarboxylase (GAD1). Results: We found that the frequencies of the HLA-DRB1*04, -DQA1*02:01, and -DQA1*03:01 alleles were significantly higher in the PD patients than in the controls, suggesting that these alleles are potential risk factors. Furthermore, the HLA-DQA1*02:01 allele was detected more frequently in the PD group when the disease onset was at 60 years or older. On the contrary, the HLA-DRB1*01 and HLA-DQA1*05:01 alleles were less common in the PD patients, indicating a possible protective effect. Regarding biomarkers, the blood levels of S100 calcium-binding protein A9 were significantly higher, and the kynurenic acid levels were significantly lower in the PD group. The NfL levels were also higher in the PD group but did not reach statistical significance, possibly due to the sensitivity limitations of the ELISA method used. The GAD1 levels showed no significant differences between the two groups. Conclusions: Our findings indicate that the HLA-DRB1*01 and -DRB1*04 alleles and the HLA-DQA1*02:01, -DQA1*03:01, and -DQA1*05:01 alleles are associated with PD. Moreover, S100 calcium-binding protein A9 and kynurenic acid can be considered potential blood biomarkers for PD. These findings contribute to the growing body of knowledge on PD and offer new directions for further research in Latvian cohorts.
Collapse
Affiliation(s)
- Olga Minibajeva
- Department of Doctoral Studies, Rīga Stradiņš University, LV-1007 Riga, Latvia
- Department of Neurology and Neurosurgery, Riga East University Hospital, LV-1079 Riga, Latvia
| | - Guntis Karelis
- Department of Neurology and Neurosurgery, Riga East University Hospital, LV-1079 Riga, Latvia
- Department of Infectology, Rīga Stradiņš University, LV-1006 Riga, Latvia
| | - Maksims Zolovs
- Statistics Unit, Rīga Stradiņš University, LV-1048 Riga, Latvia
- Institute of Life Sciences and Technology, Daugavpils University, LV-5401 Daugavpils, Latvia
| | - Viktorija Ķēniņa
- Department of Neurology, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia
- Department of Biology and Microbiology, Rīga Stradiņš University, LV-1007 Riga, Latvia
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, LV-1002 Riga, Latvia
| |
Collapse
|
20
|
Durmaz Celik N, Ozben S, Ozben T. Unveiling Parkinson's disease through biomarker research: current insights and future prospects. Crit Rev Clin Lab Sci 2024; 61:529-545. [PMID: 38529882 DOI: 10.1080/10408363.2024.2331471] [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: 09/04/2023] [Revised: 02/14/2024] [Accepted: 03/13/2024] [Indexed: 03/27/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition marked by the gradual depletion of dopaminergic neurons in the substantia nigra. Despite substantial strides in comprehending potential causative mechanisms, the validation of biomarkers with unequivocal evidence for routine clinical application remains elusive. Consequently, the diagnosis heavily relies on patients' clinical assessments and medical backgrounds. The imperative need for diagnostic and prognostic biomarkers arises due to the prevailing limitations of treatments, which predominantly address symptoms without modifying the disease course. This comprehensive review aims to elucidate the existing landscape of diagnostic and prognostic biomarkers for PD, drawing insights from contemporary literature.
Collapse
Affiliation(s)
- Nazlı Durmaz Celik
- Department of Neurology, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey
| | - Serkan Ozben
- Department of Neurology, University of Health Sciences, Antalya Training and Research Hospital, Antalya, Turkey
| | - Tomris Ozben
- Department of Medical Biochemistry, Medical Faculty, Akdeniz University, Antalya, Turkey
| |
Collapse
|
21
|
Liu J, Zhang Y. Serum neurofilament light chain: a novel biomarker for cardiovascular diseases in individuals without hypertension. Sci Rep 2024; 14:26117. [PMID: 39478121 PMCID: PMC11526128 DOI: 10.1038/s41598-024-77446-y] [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: 09/25/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Serum neurofilament light chain (sNFL) is a biomarker for axonal injury. Previous studies have linked sNFL levels to cardiovascular risk factors such as diabetes and hypertension, but its association with cardiovascular diseases (CVD) remains unclear. This study aims to explore the association between sNFL and CVD and evaluates its predictive value. Utilizing NHANES 2013-2014 data, this study included 2,035 participants aged ≥ 20 years with measured sNFL quantified using a Siemens immunoassay. CVD was self-reported and included myocardial infarction, stroke, heart failure, coronary heart disease, or angina. Logistic regression models assessed the association between sNFL levels and CVD. The predictive value of sNFL for CVD was evaluated using area under the curve (AUC) and DeLong test. Participants with higher sNFL levels were typically older, male, non-Hispanic white, smokers, and had lower socioeconomic status, higher CVD, hypertension, and diabetes prevalence. Higher sNFL levels were significantly associated with increased odds of CVD (adjusted OR = 1.41, 95% CI: 1.05-1.88). The association was significant in non-hypertensive individuals (OR = 2.72, 95% CI: 1.61-4.62) but not in hypertensive individuals (OR = 1.13, 95% CI: 0.81-1.56). sNFL addition to traditional risk models improved predictive accuracy, especially in non-hypertensive individuals (AUC from 0.827 to 0.856). sNFL levels are significantly associated with CVD in the general population, with a strong predictive value in non-hypertensive individuals. Future longitudinal studies should validate sNFL's efficacy in various populations and explore the underlying mechanisms of its relationship with hypertension and CVD.
Collapse
Affiliation(s)
- Jing Liu
- Department of Emergency Medicine, The Affiliated Second Hospital, Hengyang Medical school, University of South China, Hengyang, 421009, China
- , No. 35 Jiefang Avenue, Zhengxiang District, Hengyang City, 421001, Hunan Province, PR China
| | - Ya Zhang
- Department of Gland Surgery, The Affiliated Nanhua Hospital, Hengyang Medical school, University of South China, Hengyang, 421002, China.
| |
Collapse
|
22
|
Gonzalez-Latapi P, Gochanour C, Cho H, Ho Choi S, Caspell-Garcia C, Coffey C, Brumm M, Lafontant DE, Xiao Y, Tanner C, Venuto CS, Kieburtz K, Chahine LM, Poston KL, Siderowf A, Marek K, Simuni T. Eleven Years of Change: Disease Progression in Biomarker-Defined Sporadic Parkinson's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.09.24315191. [PMID: 39417131 PMCID: PMC11482996 DOI: 10.1101/2024.10.09.24315191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Long-term longitudinal data on outcomes in sporadic Parkinson's Disease are limited, especially from cohorts with extensive biological characterization. Recent advances in biomarkers characterization of Parkinson's Disease necessitate an updated examination of long-term progression within contemporary cohorts like the Parkinson's Progression Markers Initiative, which enrolled individuals within 2 years of clinical diagnosis of Parkinson's Disease. Our study leverages the Neuronal Synuclein Disease framework, which defines the disease based on biomarker assessed presence of neuronal alpha-synuclein and dopamine deficit, rather than based on conventional clinical diagnostic criteria. In this study we aimed to provide a comprehensive long-term description of disease progression using the integrated biological and clinical staging system framework. We analyzed data from 344 participants from the sporadic Parkinson's Disease cohort in the Parkinson's Progression Markers Initiative, who met Neuronal Synuclein Disease criteria. We assessed 11-year progression in a spectrum of clinical measures. We used Cox proportional hazards models to assess the association between baseline stage and time to key outcomes, including survival, postural instability (Hoehn & Yahr ≥ 3), loss of independence (Schwab & England < 80%), cognitive decline, and domain-based milestones such as walking and balance, motor complications, autonomic dysfunction, and activities of daily living. Additional analyses were completed to account for death and participant dropout. Biomarker analysis included dopamine transporter binding measures, as well as serum urate, neurofilament light chain and CSF amyloid-beta, phosphorylated tau and total tau. At baseline, despite the cohort consisting of individuals within 2 years of clinical diagnosis, there was clear separation of participants in Neuronal Synuclein Disease Stages (23% Stage 2b, 67% Stage 3, 10% Stage 4). At 11 years, data were available for 153 participants; 35 participants had died over the follow up period. Of retained participants, 59% presented normal cognition, 24% had evidence of postural instability and mean Schwab & England score was 78.5. Serum neurofilament light chain consistently increased over time. No other biofluids had a consistent change in trajectory. Of importance, baseline Neuronal Synuclein Disease Stage predicted progression to clinically meaningful milestones. This study provides data on longitudinal, 11-year progression in Neuronal Synuclein Disease participants within 2 years of clinical diagnosis. We observed better long-term outcomes in this contemporary observational study cohort. It highlights the heterogeneity in the early Parkinson's Disease population as defined by clinical diagnostic criteria and underscores the importance of shifting from clinical to biologically and functionally based inclusion criteria in the design of new clinical trials.
Collapse
|
23
|
Caron NS, Byrne LM, Lemarié FL, Bone JN, Aly AEE, Ko S, Anderson C, Casal LL, Hill AM, Hawellek DJ, McColgan P, Wild EJ, Leavitt BR, Hayden MR. Elevated plasma and CSF neurofilament light chain concentrations are stabilized in response to mutant huntingtin lowering in the brains of Huntington's disease mice. Transl Neurodegener 2024; 13:50. [PMID: 39380076 PMCID: PMC11460072 DOI: 10.1186/s40035-024-00443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Therapeutic approaches aimed at lowering toxic mutant huntingtin (mHTT) levels in the brain can reverse disease phenotypes in animal models of Huntington's disease (HD) and are currently being evaluated in clinical trials. Sensitive and dynamic response biomarkers are needed to assess the efficacy of such candidate therapies. Neurofilament light chain (NfL) is a biomarker of neurodegeneration that increases in cerebrospinal fluid (CSF) and blood with progression of HD. However, it remains unknown whether NfL in biofluids could serve as a response biomarker for assessing the efficacy of disease-modifying therapies for HD. METHODS Longitudinal plasma and cross-sectional CSF samples were collected from the YAC128 transgenic mouse model of HD and wild-type (WT) littermate control mice throughout the natural history of disease. Additionally, biofluids were collected from YAC128 mice following intracerebroventricular administration of an antisense oligonucleotide (ASO) targeting the mutant HTT transgene (HTT ASO), at ages both before and after the onset of disease phenotypes. NfL concentrations in plasma and CSF were quantified using ultrasensitive single-molecule array technology. RESULTS Plasma and CSF NfL concentrations were significantly elevated in YAC128 compared to WT littermate control mice from 9 months of age. Treatment of YAC128 mice with either 15 or 50 µg HTT ASO resulted in a dose-dependent, allele-selective reduction of mHTT throughout the brain at a 3-month interval, which was sustained with high-dose HTT ASO treatment for up to 6 months. Lowering of brain mHTT prior to the onset of regional brain atrophy and HD-like motor deficits in this model had minimal effect on plasma NfL at either dose, but led to a dose-dependent reduction of CSF NfL. In contrast, initiating mHTT lowering in the brain after the onset of neuropathological and behavioural phenotypes in YAC128 mice resulted in a dose-dependent stabilization of NfL increases in both plasma and CSF. CONCLUSIONS Our data provide evidence that the response of NfL in biofluids is influenced by the magnitude of mHTT lowering in the brain and the timing of intervention, suggesting that NfL may serve as a promising exploratory response biomarker for HD.
Collapse
Affiliation(s)
- Nicholas S Caron
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Lauren M Byrne
- UCL Huntington's Disease Centre, University College London Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Fanny L Lemarié
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Jeffrey N Bone
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Statistics, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
| | - Amirah E-E Aly
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Seunghyun Ko
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Christine Anderson
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Lorenzo L Casal
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Austin M Hill
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - David J Hawellek
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Peter McColgan
- Roche Products Ltd., Welwyn Garden City, AL7 1TW, United Kingdom
| | - Edward J Wild
- UCL Huntington's Disease Centre, University College London Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
24
|
Lin T, Mao H, Huang S, Chen J. Association between aromatic amines and serum neurofilament light chain as a biomarker of neural damage: a cross-sectional study from NHANES. Front Public Health 2024; 12:1344087. [PMID: 39381758 PMCID: PMC11458512 DOI: 10.3389/fpubh.2024.1344087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Background Aromatic amines (AAs) are a group of compounds widely found in chemical industry, tobacco smoke, and during food processing, with established carcinogenic properties. To date, there have been no reports on the potential neurotoxic effects of adult exposure to AAs. Serum neurofilament light chain (sNfL) is a protein released into the bloodstream following nerve axon injury and has been validated as a reliable biomarker for various neurological diseases. However, there has been no research to investigate the relationship between AAs exposure and sNfL. Methods In this study, we selected adults (aged ≥20 years) with data on both AAs and sNfL from the National Health and Nutrition Examination Survey (NHANES) conducted in 2013-2014. We used multivariable linear regression models to explore the correlation between urinary AAs and sNfL. Results In total, 510 adult participants with an average age of 43.58 ± 14.74 years were included in the study. Our findings indicate that, based on univariate linear regression and between-group comparative analyses, 1-Aminonaphthalene (1-AN), 2-Aminonaphthalene (2-AN), 4-Aminobiphenyl (4-AN) and o-Anisidine (o-ANI) showed a positive correlation with serum neurofilament light chain (P < 0.05). However, multiple linear regression analysis revealed that only 2-AN exhibited a positive correlation with serum neurofilament light chain (P < 0.05), while the correlations of other compounds with serum neurofilament light chain became non-significant. Conclusion Although our cross-sectional study fails to establish causal relationships or determine clinical significance, the findings indicate a potential association between adult exposure to AAs, notably 2-AN, and nerve damage. Consequently, further research is needed to explore the connection between AAs exposure, sNfL, and neurological conditions in adults.
Collapse
Affiliation(s)
- Tong Lin
- Department of Critical Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | | | | | | |
Collapse
|
25
|
Kasanga EA, Soto I, Centner A, McManus R, Shifflet MK, Navarrete W, Han Y, Lisk J, Ehrhardt T, Wheeler K, Mhatre-Winters I, Richardson JR, Bishop C, Nejtek VA, Salvatore MF. Moderate intensity aerobic exercise alleviates motor deficits in 6-OHDA lesioned rats and reduces serum levels of biomarkers of Parkinson's disease severity without recovery of striatal dopamine or tyrosine hydroxylase. Exp Neurol 2024; 379:114875. [PMID: 38944332 DOI: 10.1016/j.expneurol.2024.114875] [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: 03/20/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
Alleviation of motor impairment by aerobic exercise (AE) in Parkinson's disease (PD) patients points to activation of neurobiological mechanisms that may be targetable by therapeutic approaches. However, evidence for AE-related recovery of striatal dopamine (DA) signaling or tyrosine hydroxylase (TH) loss has been inconsistent in rodent studies. This ambiguity may be related to the timing of AE intervention in relation to the status of nigrostriatal neuron loss. Here, we replicated human PD at diagnosis by establishing motor impairment with >80% striatal DA and TH loss prior to initiating AE, and assessed its potential to alleviate motor decline and restore DA and TH loss. We also evaluated if serum levels of neurofilament light (NfL) and glial fibrillary acidic protein (GFAP), biomarkers of human PD severity, changed in response to AE. 6-hydroxydopamine (6-OHDA) was infused unilaterally into rat medial forebrain bundle to induce progressive nigrostriatal neuron loss over 28 days. Moderate intensity AE (3× per week, 40 min/session), began 8-10 days post-lesion following establishment of impaired forelimb use. Striatal tissue DA, TH protein and mRNA, and serum levels of NfL/GFAP were determined 3-wks after AE began. Despite severe striatal DA depletion at AE initiation, forelimb use deficits and hypokinesia onset were alleviated by AE, without recovery of striatal DA or TH protein loss, but reduced NfL and GFAP serum levels. This proof-of-concept study shows AE alleviates motor impairment when initiated with >80% striatal DA loss without obligate recovery of striatal DA or TH protein. Moreover, the AE-related reduction of NfL and GFAP serum levels may serve as objective blood-based biomarkers of AE efficacy.
Collapse
Affiliation(s)
- Ella A Kasanga
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Isabel Soto
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Ashley Centner
- Department of Psychology, Binghamton University, Binghamton, NY, United States of America
| | - Robert McManus
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Marla K Shifflet
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Walter Navarrete
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Jerome Lisk
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Travis Ehrhardt
- Clearcut Ortho Rehab & Diagnostics, Fort Worth, TX, United States of America
| | - Ken Wheeler
- Clearcut Ortho Rehab & Diagnostics, Fort Worth, TX, United States of America
| | - Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Jason R Richardson
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, NY, United States of America
| | - Vicki A Nejtek
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Michael F Salvatore
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America.
| |
Collapse
|
26
|
Bavato F, Barro C, Schnider LK, Simrén J, Zetterberg H, Seifritz E, Quednow BB. Introducing neurofilament light chain measure in psychiatry: current evidence, opportunities, and pitfalls. Mol Psychiatry 2024; 29:2543-2559. [PMID: 38503931 PMCID: PMC11412913 DOI: 10.1038/s41380-024-02524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
The recent introduction of new-generation immunoassay methods allows the reliable quantification of structural brain markers in peripheral matrices. Neurofilament light chain (NfL), a neuron-specific cytoskeletal component released in extracellular matrices after neuroaxonal impairment, is considered a promising blood marker of active brain pathology. Given its sensitivity to a wide range of neuropathological alterations, NfL has been suggested for the use in clinical practice as a highly sensitive, but unspecific tool to quantify active brain pathology. While large efforts have been put in characterizing its clinical profile in many neurological conditions, NfL has received far less attention as a potential biomarker in major psychiatric disorders. Therefore, we briefly introduce NfL as a marker of neuroaxonal injury, systematically review recent findings on cerebrospinal fluid and blood NfL levels in patients with primary psychiatric conditions and highlight the opportunities and pitfalls. Current evidence suggests an elevation of blood NfL levels in patients with major depression, bipolar disorder, psychotic disorders, anorexia nervosa, and substance use disorders compared to physiological states. However, blood NfL levels strongly vary across diagnostic entities, clinical stage, and patient subgroups, and are influenced by several demographic, clinical, and analytical factors, which require accurate characterization. Potential clinical applications of NfL measure in psychiatry are seen in diagnostic and prognostic algorithms, to exclude neurodegenerative disease, in the assessment of brain toxicity for different pharmacological compounds, and in the longitudinal monitoring of treatment response. The high inter-individual variability of NfL levels and the lack of neurobiological understanding of its release are some of the main current limitations. Overall, this primer aims to introduce researchers and clinicians to NfL measure in the psychiatric field and to provide a conceptual framework for future research directions.
Collapse
Affiliation(s)
- Francesco Bavato
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Christian Barro
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura K Schnider
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy and Psychosomatics; Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Chahine LM, Lafontant DE, Ho Choi S, Iwaki H, Blauwendraat C, Singleton AB, Brumm MC, Alcalay RN, Merchant K, Nudelman KNH, Dagher A, Vo A, Tao Q, Venuto CS, Kieburtz K, Poston KL, Bressman S, Gonzalez-Latapi P, Avants B, Coffey C, Jennings D, Tolosa E, Siderowf A, Marek K, Simuni T. LRRK2-Associated Parkinsonism With and Without In Vivo Evidence of Alpha-Synuclein Aggregates. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.22.24310806. [PMID: 39108519 PMCID: PMC11302724 DOI: 10.1101/2024.07.22.24310806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Among LRRK2-associated parkinsonism cases with nigral degeneration, over two-thirds demonstrate evidence of pathologic alpha-synuclein, but many do not. Understanding the clinical phenotype and underlying biology in such individuals is critical for therapeutic development. Our objective was to compare clinical and biomarker features, and rate of progression over 4 years follow-up, among LRRK2-associated parkinsonism cases with and without in vivo evidence of alpha-synuclein aggregates. Methods Data were from the Parkinson's Progression Markers Initiative, a multicenter prospective cohort study. The sample included individuals diagnosed with Parkinson disease with pathogenic variants in LRRK2. Presence of CSF alpha-synuclein aggregation was assessed with seed amplification assay. A range of clinician- and patient- reported outcome assessments were administered. Biomarkers included dopamine transporter SPECT scan, CSF amyloid-beta1-42, total tau, phospho-tau181, urine bis(monoacylglycerol)phosphate levels, and serum neurofilament light chain. Linear mixed effects models examined differences in trajectory in CSF negative and positive groups. Results 148 LRRK2-parkinsonism cases (86% with G2019S variant), 46 negative and 102 positive for CSF alpha-synuclein seed amplification assay were included. At baseline, the negative group were older than the positive group (median [interquartile range] 69.1 [65.2-72.3] vs 61.5 [55.6-66.9] years, p<0.001) and a greater proportion were female (28 (61%) vs 43 (42%), p=0.035). Despite being older, the negative group had similar duration since diagnosis, and similar motor rating scale (16 [11-23] vs 16 [10-22], p=0.480) though lower levodopa equivalents. Only 13 (29%) of the negative group were hyposmic, compared to 75 (77%) of the positive group. Lowest putamen dopamine transporter binding expected for age and sex was greater in the negative vs positive groups (0.36 [0.29-0.45] vs 0.26 [0.22-0.37], p<0.001). Serum neurofilament light chain was higher in the negative group compared to the positive group (17.10 [13.60-22.10] vs 10.50 [8.43-14.70]; age-adjusted p-value=0.013). In terms of longitudinal change, the negative group remained stable in functional rating scale score in contrast to the positive group who had a significant increase (worsening) of 0.729 per year (p=0.037), but no other differences in trajectory were found. Conclusion Among individuals diagnosed with Parkinson disease with pathogenic variants in the LRRK2 gene, we found clinical and biomarker differences in cases without versus with in vivo evidence of CSF alpha-synuclein aggregates. LRRK2 parkinsonism cases without evidence of alpha-synuclein aggregates as a group exhibit less severe motor manifestations and decline may have more significant cognitive dysfunction. The underlying biology in LRRK2-parkinsonism cases without evidence of alpha-synuclein aggregates requires further investigation.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - David-Erick Lafontant
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Seung Ho Choi
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Hirotaka Iwaki
- DataTecnica LLC, Washington, District of Columbia, USA. (2) Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael C Brumm
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Roy N Alcalay
- Neurological Institute, Tel-Aviv Medical Center, Tel-Aviv, Israel and Department of Neurology; Columbia University Irving Medical Center
| | - Kalpana Merchant
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Andrew Vo
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Qin Tao
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Charles S Venuto
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY
| | - Karl Kieburtz
- Department of Neurology, Center for Health and Technology, University of Rochester Medical Center, Rochester, NY
| | - Kathleen L Poston
- Department of Neurology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Susan Bressman
- Department of Neurology, Mount Sinai Beth Israel and Icahn School of Medicine, Mount Sinai, New York City, New York, USA
| | - Paulina Gonzalez-Latapi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Christopher Coffey
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | | | - Eduard Tolosa
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII) Barcelona, Spain
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ken Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
28
|
Mousele C, Holden D, Gnanapavan S. Neurofilaments in neurologic disease. Adv Clin Chem 2024; 123:65-128. [PMID: 39181624 DOI: 10.1016/bs.acc.2024.06.010] [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: 08/27/2024]
Abstract
Neurofilaments (NFs), major cytoskeletal constituents of neurons, have emerged as universal biomarkers of neuronal injury. Neuroaxonal damage underlies permanent disability in various neurological conditions. It is crucial to accurately quantify and longitudinally monitor this damage to evaluate disease progression, evaluate treatment effectiveness, contribute to novel treatment development, and offer prognostic insights. Neurofilaments show promise for this purpose, as their levels increase with neuroaxonal damage in both cerebrospinal fluid and blood, independent of specific causal pathways. New assays with high sensitivity allow reliable measurement of neurofilaments in body fluids and open avenues to investigate their role in neurological disorders. This book chapter will delve into the evolving landscape of neurofilaments, starting with their structure and cellular functions within neurons. It will then provide a comprehensive overview of their broad clinical value as biomarkers in diseases affecting the central or peripheral nervous system.
Collapse
|
29
|
Couto B, Sousa M, Gonzalez-Latapi P, McArthur E, Lang A, Chen-Plotkin A, Marras C. Disease Progression and Sphingolipids and Neurofilament Light Chain in Early Idiopathic Parkinson's Disease. Can J Neurol Sci 2024; 51:573-576. [PMID: 37641969 DOI: 10.1017/cjn.2023.281] [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: 08/31/2023]
Abstract
Parkinson's disease(PD) lacks a biomarker for disease progression. To analyze how cerebrospinal fluid (CSF), glucosylceramide (GlcCer), sphingomyelin (SM), or serum neurofilament light chain (NfL) associate with progression of PD in a retrospective cohort, we used linear mixed-model regressions between baseline biomarkers and change in dopamine transporter brain-imaging (DaTscan©), Montreal cognitive assesment (MoCA), or global composite outcome (GCO) score. In 191 PD patients, biomarkers were not associated with DaTscan or MoCA change over 2.1 years. Higher baseline GlcCer/SM ratio and serum-NfL nonsignificantly associated with increase in GCO score. Results do not support a role for CSF-sphingolipid/serum-NfL to predict cognitive and DaTscan progression in early-PD. Potential prediction of global clinical change warrants further study.
Collapse
Affiliation(s)
- Blas Couto
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
- Institute of Cognitive and Traslational Neuroscience (INCyT), at the INECO-CONICET-Favaloro University Hospital, Buenos Aires, Argentina
| | - Mario Sousa
- Department of Neurology, Inselspital, Bern University Hospital, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Paulina Gonzalez-Latapi
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | - Anthony Lang
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Connie Marras
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
30
|
Hällqvist J, Bartl M, Dakna M, Schade S, Garagnani P, Bacalini MG, Pirazzini C, Bhatia K, Schreglmann S, Xylaki M, Weber S, Ernst M, Muntean ML, Sixel-Döring F, Franceschi C, Doykov I, Śpiewak J, Vinette H, Trenkwalder C, Heywood WE, Mills K, Mollenhauer B. Plasma proteomics identify biomarkers predicting Parkinson's disease up to 7 years before symptom onset. Nat Commun 2024; 15:4759. [PMID: 38890280 PMCID: PMC11189460 DOI: 10.1038/s41467-024-48961-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Parkinson's disease is increasingly prevalent. It progresses from the pre-motor stage (characterised by non-motor symptoms like REM sleep behaviour disorder), to the disabling motor stage. We need objective biomarkers for early/pre-motor disease stages to be able to intervene and slow the underlying neurodegenerative process. Here, we validate a targeted multiplexed mass spectrometry assay for blood samples from recently diagnosed motor Parkinson's patients (n = 99), pre-motor individuals with isolated REM sleep behaviour disorder (two cohorts: n = 18 and n = 54 longitudinally), and healthy controls (n = 36). Our machine-learning model accurately identifies all Parkinson patients and classifies 79% of the pre-motor individuals up to 7 years before motor onset by analysing the expression of eight proteins-Granulin precursor, Mannan-binding-lectin-serine-peptidase-2, Endoplasmatic-reticulum-chaperone-BiP, Prostaglaindin-H2-D-isomaerase, Interceullular-adhesion-molecule-1, Complement C3, Dickkopf-WNT-signalling pathway-inhibitor-3, and Plasma-protease-C1-inhibitor. Many of these biomarkers correlate with symptom severity. This specific blood panel indicates molecular events in early stages and could help identify at-risk participants for clinical trials aimed at slowing/preventing motor Parkinson's disease.
Collapse
Affiliation(s)
- Jenny Hällqvist
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK.
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK.
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany.
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany.
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Chiara Pirazzini
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Kailash Bhatia
- National Hospital for Neurology & Neurosurgery, Queen Square, WC1N3BG, London, UK
| | | | - Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Marielle Ernst
- Institute of Diagnostic and Interventional Neuroradiology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Claudio Franceschi
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Ivan Doykov
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Justyna Śpiewak
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Héloїse Vinette
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
- UCL: Food, Microbiomes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Wendy E Heywood
- UCL Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Kevin Mills
- UCL Queen Square Institute of Neurology, Clinical and Movement Neurosciences, London, UK
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
31
|
Zhao J, Zhao P. Association between serum neurofilament light chain and periodontitis. Clin Oral Investig 2024; 28:369. [PMID: 38864919 PMCID: PMC11168977 DOI: 10.1007/s00784-024-05769-1] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVES The association between serum neurofilament light chain (sNfL) and periodontitis remains unclear, and there is a need to examine the contribution of serum albumin (SA) in this association. The objective of the study is to investigate the correlation between sNfLand periodontitis, while examining the potential mediator role of SA in this association. METHODS The study, which included 1218 participants from the 2013-2014 National Health and Nutrition Examination Survey (NHANES), aimed to evaluate the association between sNfL and periodontitis through weighted multivariable logistic regression analysis, restricted cubic spline (RCS) models, and stratified models. In addition, mediation analysis was used to investigate the role of SA in mediating this association. RESULTS The multivariable logistic regression models revealed that sNfL was significantly linked to periodontitis (model 1: odds ratio [OR], 3.08, 95% confidence interval [CI], 1.48 to 6.39, model 2: OR, 3.69; 95% CI, 1.73 to 7.90, model 3: OR, 3.58, 95% CI, 1.52 to 8.43). The RCS models suggested a linear relationship between sNfL and periodontitis. The stratified analysis revealed no significant moderating effects (p-value > 0.05). The mediation analysis demonstrated that SA mediated the correlation between sNfL and periodontitis, with a mediation proportion of 10.62%. CONCLUSIONS The results point to sNfL being a factor in the heightened risk of periodontitis. Additionally, SA may mediate the changes in periodontitis that are associated with sNfL. CLINICAL RELEVANCE sNfL may contribute to the development of periodontitis by mediating changes in SA in humans.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Xindu Road 606#, Yancheng, Jiangsu Province, PR China.
| | - Panwen Zhao
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Xindu Road 606#, Yancheng, Jiangsu Province, PR China
| |
Collapse
|
32
|
Bartl M, Nilsson J, Dakna M, Weber S, Schade S, Xylaki M, Fernandes Gomes B, Ernst M, Muntean ML, Sixel-Döring F, Trenkwalder C, Zetterberg H, Brinkmalm A, Mollenhauer B. Lysosomal and synaptic dysfunction markers in longitudinal cerebrospinal fluid of de novo Parkinson's disease. NPJ Parkinsons Dis 2024; 10:102. [PMID: 38760408 PMCID: PMC11101466 DOI: 10.1038/s41531-024-00714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
Lysosomal and synaptic dysfunctions are hallmarks in neurodegeneration and potentially relevant as biomarkers, but data on early Parkinson's disease (PD) is lacking. We performed targeted mass spectrometry with an established protein panel, assessing autophagy and synaptic function in cerebrospinal fluid (CSF) of drug-naïve de novo PD, and sex-/age-matched healthy controls (HC) cross-sectionally (88 PD, 46 HC) and longitudinally (104 PD, 58 HC) over 10 years. Multiple markers of autophagy, synaptic plasticity, and secretory pathways were reduced in PD. We added samples from prodromal subjects (9 cross-sectional, 12 longitudinal) with isolated REM sleep behavior disorder, revealing secretogranin-2 already decreased compared to controls. Machine learning identified neuronal pentraxin receptor and neurosecretory protein VGF as most relevant for discriminating between groups. CSF levels of LAMP2, neuronal pentraxins, and syntaxins in PD correlated with clinical progression, showing predictive potential for motor- and non-motor symptoms as a valid basis for future drug trials.
Collapse
Affiliation(s)
- Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany.
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany.
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Sandrina Weber
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Mary Xylaki
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Bárbara Fernandes Gomes
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Marielle Ernst
- Institute of Diagnostic and Interventional Neuroradiology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Ann Brinkmalm
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| |
Collapse
|
33
|
Khalil M, Teunissen CE, Lehmann S, Otto M, Piehl F, Ziemssen T, Bittner S, Sormani MP, Gattringer T, Abu-Rumeileh S, Thebault S, Abdelhak A, Green A, Benkert P, Kappos L, Comabella M, Tumani H, Freedman MS, Petzold A, Blennow K, Zetterberg H, Leppert D, Kuhle J. Neurofilaments as biomarkers in neurological disorders - towards clinical application. Nat Rev Neurol 2024; 20:269-287. [PMID: 38609644 DOI: 10.1038/s41582-024-00955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
Neurofilament proteins have been validated as specific body fluid biomarkers of neuro-axonal injury. The advent of highly sensitive analytical platforms that enable reliable quantification of neurofilaments in blood samples and simplify longitudinal follow-up has paved the way for the development of neurofilaments as a biomarker in clinical practice. Potential applications include assessment of disease activity, monitoring of treatment responses, and determining prognosis in many acute and chronic neurological disorders as well as their use as an outcome measure in trials of novel therapies. Progress has now moved the measurement of neurofilaments to the doorstep of routine clinical practice for the evaluation of individuals. In this Review, we first outline current knowledge on the structure and function of neurofilaments. We then discuss analytical and statistical approaches and challenges in determining neurofilament levels in different clinical contexts and assess the implications of neurofilament light chain (NfL) levels in normal ageing and the confounding factors that need to be considered when interpreting NfL measures. In addition, we summarize the current value and potential clinical applications of neurofilaments as a biomarker of neuro-axonal damage in a range of neurological disorders, including multiple sclerosis, Alzheimer disease, frontotemporal dementia, amyotrophic lateral sclerosis, stroke and cerebrovascular disease, traumatic brain injury, and Parkinson disease. We also consider the steps needed to complete the translation of neurofilaments from the laboratory to the management of neurological diseases in clinical practice.
Collapse
Affiliation(s)
- Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maria Pia Sormani
- Department of Health Sciences, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Thomas Gattringer
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Thebault
- Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdelhak
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Ari Green
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Pascal Benkert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Ludwig Kappos
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Manuel Comabella
- Neurology Department, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hayrettin Tumani
- Department of Neurology, CSF Laboratory, Ulm University Hospital, Ulm, Germany
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Axel Petzold
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurology, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
- Moorfields Eye Hospital, The National Hospital for Neurology and Neurosurgery and the Queen Square Institute of Neurology, UCL, London, UK
| | - Kaj Blennow
- 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
- 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
- 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
- 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, WI, USA
| | - David Leppert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland.
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
34
|
Janssen Daalen JM, Gerritsen A, Gerritse G, Gouman J, Meijerink H, Rietdijk LE, Darweesh SKL. How Lifetime Evolution of Parkinson's Disease Could Shape Clinical Trial Design: A Shared Patient-Clinician Viewpoint. Brain Sci 2024; 14:358. [PMID: 38672010 PMCID: PMC11048137 DOI: 10.3390/brainsci14040358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Parkinson's disease (PD) has a long, heterogeneous, pre-diagnostic phase, during which pathology insidiously accumulates. Increasing evidence suggests that environmental and lifestyle factors in early life contribute to disease risk and progression. Thanks to the extensive study of this pre-diagnostic phase, the first prevention trials of PD are being designed. However, the highly heterogenous evolution of the disease across the life course is not yet sufficiently taken into account. This could hamper clinical trial success in the advent of biological disease definitions. In an interdisciplinary patient-clinician study group, we discussed how an approach that incorporates the lifetime evolution of PD may benefit the design of disease-modifying trials by impacting population, target and outcome selection. We argue that the timepoint of exposure to risk and protective factors plays a critical role in PD subtypes, influencing population selection. In addition, recent developments in differential disease mechanisms, aided by biological disease definitions, could impact optimal treatment targets. Finally, multimodal biomarker panels using this lifetime approach will likely be most sensitive as progression markers for more personalized trials. We believe that the lifetime evolution of PD should be considered in the design of clinical trials, and that such initiatives could benefit from more patient-clinician partnerships.
Collapse
Affiliation(s)
- Jules M. Janssen Daalen
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, 6525 GA Nijmegen, The Netherlands; (J.M.J.D.); (A.G.)
| | - Aranka Gerritsen
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, 6525 GA Nijmegen, The Netherlands; (J.M.J.D.); (A.G.)
| | - Gijs Gerritse
- Dutch Parkinson’s Patient Association, P.O. Box 46, 3980 CA Bunnik, The Netherlands; (G.G.); (J.G.); (H.M.); (L.E.R.)
| | - Jan Gouman
- Dutch Parkinson’s Patient Association, P.O. Box 46, 3980 CA Bunnik, The Netherlands; (G.G.); (J.G.); (H.M.); (L.E.R.)
| | - Hannie Meijerink
- Dutch Parkinson’s Patient Association, P.O. Box 46, 3980 CA Bunnik, The Netherlands; (G.G.); (J.G.); (H.M.); (L.E.R.)
| | - Leny E. Rietdijk
- Dutch Parkinson’s Patient Association, P.O. Box 46, 3980 CA Bunnik, The Netherlands; (G.G.); (J.G.); (H.M.); (L.E.R.)
| | - Sirwan K. L. Darweesh
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, 6525 GA Nijmegen, The Netherlands; (J.M.J.D.); (A.G.)
| |
Collapse
|
35
|
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
|
36
|
Wang X, Dong T, Li X, Yu W, Jia Z, Liu Y, Yang J. Global biomarker trends in Parkinson's disease research: A bibliometric analysis. Heliyon 2024; 10:e27437. [PMID: 38501016 PMCID: PMC10945172 DOI: 10.1016/j.heliyon.2024.e27437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
As the second most common neurodegenerative disease globally, Parkinson's disease (PD) affects millions of people worldwide. In recent years, the scientific publications related to PD biomarker research have exploded, reflecting the growing interest in unraveling the complex pathophysiology of PD. In this study, we aim to use various bibliometric tools to identify key scientific concepts, detect emerging trends, and analyze the global trends and development of PD biomarker research.The research encompasses various stages of biomarker development, including exploration, identification, and multi-modal research. MOVEMENT DISORDERS emerged as the leading journal in terms of publications and citations. Key authors such as Mollenhauer and Salem were identified, while the University of Pennsylvania and USA stood out in collaboration and research output. NEUROSCIENCES emerged as the most important research direction. Key biomarker categories include α-synuclein-related markers, neurotransmitter-related markers, inflammation and immune system-related markers, oxidative stress and mitochondrial function-related markers, and brain imaging-related markers. Furthermore, future trends in PD biomarker research focus on exosomes and plasma biomarkers, miRNA, cerebrospinal fluid biomarkers, machine learning applications, and animal models of PD. These trends contribute to early diagnosis, disease progression monitoring, and understanding the pathological mechanisms of PD.
Collapse
Affiliation(s)
- Xingxin Wang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tiantian Dong
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xuhao Li
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenyan Yu
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhixia Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| |
Collapse
|
37
|
Wang X, Dong T, Li X, Yu W, Jia Z, Liu Y, Yang J. Global biomarker trends in Parkinson's disease research: A bibliometric analysis. Heliyon 2024; 10:e27437. [PMID: 38501016 DOI: 10.1016/j.heliyon.2024.e27437if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 02/28/2024] [Indexed: 07/25/2024] Open
Abstract
As the second most common neurodegenerative disease globally, Parkinson's disease (PD) affects millions of people worldwide. In recent years, the scientific publications related to PD biomarker research have exploded, reflecting the growing interest in unraveling the complex pathophysiology of PD. In this study, we aim to use various bibliometric tools to identify key scientific concepts, detect emerging trends, and analyze the global trends and development of PD biomarker research.The research encompasses various stages of biomarker development, including exploration, identification, and multi-modal research. MOVEMENT DISORDERS emerged as the leading journal in terms of publications and citations. Key authors such as Mollenhauer and Salem were identified, while the University of Pennsylvania and USA stood out in collaboration and research output. NEUROSCIENCES emerged as the most important research direction. Key biomarker categories include α-synuclein-related markers, neurotransmitter-related markers, inflammation and immune system-related markers, oxidative stress and mitochondrial function-related markers, and brain imaging-related markers. Furthermore, future trends in PD biomarker research focus on exosomes and plasma biomarkers, miRNA, cerebrospinal fluid biomarkers, machine learning applications, and animal models of PD. These trends contribute to early diagnosis, disease progression monitoring, and understanding the pathological mechanisms of PD.
Collapse
Affiliation(s)
- Xingxin Wang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tiantian Dong
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xuhao Li
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenyan Yu
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhixia Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| |
Collapse
|
38
|
Hutchison RM, Fraser K, Yang M, Fox T, Hirschhorn E, Njingti E, Scott D, Bedell BJ, Kistner KM, Cedarbaum JM, Evans KC, Graham D, Martarello L, Mollenhauer B, Lang AE, Dam T, Beaver J. Cinpanemab in Early Parkinson Disease: Evaluation of Biomarker Results From the Phase 2 SPARK Clinical Trial. Neurology 2024; 102:e209137. [PMID: 38315945 DOI: 10.1212/wnl.0000000000209137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/29/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Sensitive, reliable, and scalable biomarkers are needed to accelerate the development of therapies for Parkinson disease (PD). In this study, we evaluate the biomarkers of early PD diagnosis, disease progression, and treatment effect collected in the SPARK. METHODS Cinpanemab is a human-derived monoclonal antibody binding preferentially to aggregated forms of extracellular α-synuclein. SPARK was a randomized, double-blind, placebo-controlled, phase 2 multicenter trial evaluating 3 cinpanemab doses administered intravenously every 4 weeks for 52 weeks with an active treatment dose-blind extension period for up to 112 weeks. SPARK enrolled 357 participants diagnosed with PD within 3 years, aged 40-80 years, ≤2.5 on the modified Hoehn and Yahr scale, and with evidence of striatal dopaminergic deficit. The primary outcome was change from baseline in the Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale total score. Secondary and exploratory biomarker outcomes evaluated change from baseline at week 52 relative to placebo. Dopamine transporter SPECT and MRI were used to quantify changes in the nigrostriatal dopamine pathway and regional atrophy. CSF and plasma samples were used to assess change in total α-synuclein levels, α-synuclein seeding, and neurofilament light chain levels. SPARK was conducted from January 2018 to April 2021 and terminated due to lack of efficacy. RESULTS Approximately 3.8% (15/398) of SPECT-imaged participants did not have evidence of dopaminergic deficit and were screen-failed. Binary classification of α-synuclein seeding designated 93% (110/118) of the enrolled CSF subgroup as positive for α-synuclein seeds at baseline. Clinical disease progression was observed, with no statistically significant difference in cinpanemab groups compared with that in placebo. Ninety-nine percent of participants with positive α-synuclein seeding remained positive through week 52. No statistically significant changes from baseline were observed between treatment groups and placebo across biomarker measures. Broadly, there was minimal annual change with high interindividual variability across biomarkers-with striatal binding ratios of the ipsilateral putamen showing the greatest mean change/SD over time. DISCUSSION Biomarker results indicated enrollment of the intended population with early PD, but there was no significant correlation with disease progression or clear evidence of a cinpanemab treatment effect on biomarker measures. Suitable biomarkers for evaluating disease severity and progression in early PD trials are still needed. TRIAL REGISTRATION INFORMATION NCT03318523 (clinicaltrials.gov/ct2/show/NCT03318523); Submitted October 24, 2017; First patient enrolled January 2018.
Collapse
Affiliation(s)
- R Matthew Hutchison
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Kyle Fraser
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Minhua Yang
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Tara Fox
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Elizabeth Hirschhorn
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Edwin Njingti
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - David Scott
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Barry J Bedell
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Kristi M Kistner
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Jesse M Cedarbaum
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Karleyton C Evans
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Danielle Graham
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Laurent Martarello
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Brit Mollenhauer
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Anthony E Lang
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - Tien Dam
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| | - John Beaver
- From Biogen Inc. (R.M.H., K.F., M.Y., E.H., K.C.E., D.G., L.M., J.B.), Cambridge, MA; Biogen Inc. (T.F.), Maidenhead, United Kingdom; Formerly Biogen Inc. at time of study (E.N., J.M.C., T.D.); Clario (D.S.), Princeton, NJ; Biospective Inc. (B.J.B.), Montreal, Quebec, Canada; Nucleus Global (K.M.K.), Atlanta, GA; Coeruleus Clinical Sciences LLC (J.M.C.), Woodbridge, CT; Department of Neurology (B.M.), University Medical Center, Göttingen and Paracelsus-Elena-Klinik, Kassel, and Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Germany; Morton and Gloria Shulman Movement Disorders Clinic (A.E.L.); and Edmond J. Safra Program in Parkinson's Disease (A.E.L.), Toronto, Ontario, Canada
| |
Collapse
|
39
|
Yin P, Niu X, Guan C, Zhang Z, Liu Y, Li J, Cui G, Zan K, Xu C. Relationship between increased serum neurofilament light chain and glial fibrillary acidic protein levels with non-motor symptoms in patients with Parkinson's disease. Psychogeriatrics 2024; 24:415-425. [PMID: 38339819 DOI: 10.1111/psyg.13088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND This study set out to investigate the relationship between serum neurofilament light chain (NFL), glial fibrillary acidic protein (GFAP), and various non-motor symptoms (NMSs) in patients with Parkinson's disease (PD). METHODS The study included 37 healthy controls (HCs) and 51 PD patients. Clinical assessments of PD symptoms were conducted for all PD patients. The NMSS was utilised to evaluate the NMS burden (NMSB) in individuals. Based on the severity of NMSB, we further categorised the PD group into two subgroups: mild-moderate NMSB group and severe-very severe NMSB group. The amounts of NFL and GFAP in the serum were measured using an extremely sensitive single molecule array (Simoa) method. Statistical analyses were performed on the collected data using SPSS 26.0 and R (version 3.6.3). RESULTS Serum GFAP and NFL levels in the PD group with severe-very severe NMSB were significantly higher than those in the mild-moderate NMSB group (GFAP: P < 0.007; NFL: P < 0.009). Serum NFL and GFAP levels had positive correlations with NMSS total scores (GFAP: r = 0.326, P = 0.020; NFL: r = 0.318, P = 0.023) and multiple subdomains. The relationship between the attention/memory domains of NMSS and NFL levels is significantly positive (r = 0.283, P = 0.044). Similarly, the mood/apathy domains of NMSS are also significantly positively correlated with GFAP levels (r = 0.441, P = 0.001). Patients with emotional problems or cognitive impairment had higher GFAP or NFL levels, respectively. Furthermore, it has been demonstrated that NMSs play a mediating role in the quality of life of patients with PD. Moreover, the combination of NFL and GFAP has proven to be more effective than using a single component in identifying PD patients with severe-very severe NMSB. CONCLUSIONS The severity of NMSs in PD patients, particularly cognitive and emotional symptoms, was found to be associated with the levels of serum NFL and GFAP. This study marks the first attempt to examine the connection between NMSs of PD and the simultaneous identification of NFL and GFAP levels.
Collapse
Affiliation(s)
- Peixiao Yin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Xuebin Niu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Chenyang Guan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Zixuan Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Yuning Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Jinyu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Kun Zan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Chuanying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The First Clinical College, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
40
|
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
|
41
|
Cousins KAQ, Irwin DJ, Tropea TF, Rhodes E, Phillips J, Chen-Plotkin AS, Brumm MC, Coffey CS, Kang JH, Simuni T, Foroud TM, Toga AW, Tanner CM, Kieburtz KD, Mollenhauer B, Galasko D, Hutten S, Weintraub D, Siderowf AD, Marek K, Poston KL, Shaw LM. Evaluation of ATN PD Framework and Biofluid Markers to Predict Cognitive Decline in Early Parkinson Disease. Neurology 2024; 102:e208033. [PMID: 38306599 PMCID: PMC11383879 DOI: 10.1212/wnl.0000000000208033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/13/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In Parkinson disease (PD), Alzheimer disease (AD) copathology is common and clinically relevant. However, the longitudinal progression of AD CSF biomarkers-β-amyloid 1-42 (Aβ42), phosphorylated tau 181 (p-tau181), and total tau (t-tau)-in PD is poorly understood and may be distinct from clinical AD. Moreover, it is unclear whether CSF p-tau181 and serum neurofilament light (NfL) have added prognostic utility in PD, when combined with CSF Aβ42. First, we describe longitudinal trajectories of biofluid markers in PD. Second, we modified the AD β-amyloid/tau/neurodegeneration (ATN) framework for application in PD (ATNPD) using CSF Aβ42 (A), p-tau181 (T), and serum NfL (N) and tested ATNPD prediction of longitudinal cognitive decline in PD. METHODS Participants were selected from the Parkinson's Progression Markers Initiative cohort, clinically diagnosed with sporadic PD or as controls, and followed up annually for 5 years. Linear mixed-effects models (LMEMs) tested the interaction of diagnosis with longitudinal trajectories of analytes (log transformed, false discovery rate [FDR] corrected). In patients with PD, LMEMs tested how baseline ATNPD status (AD [A+T+N±] vs not) predicted clinical outcomes, including Montreal Cognitive Assessment (MoCA; rank transformed, FDR corrected). RESULTS Participants were 364 patients with PD and 168 controls, with comparable baseline mean (±SD) age (patients with PD = 62 ± 10 years; controls = 61 ± 11 years]; Mann-Whitney Wilcoxon: p = 0.4) and sex distribution (patients with PD = 231 male individuals [63%]; controls = 107 male individuals [64%]; χ2: p = 1). Patients with PD had overall lower CSF p-tau181 (β = -0.16, 95% CI -0.23 to -0.092, p = 2.2e-05) and t-tau than controls (β = -0.13, 95% CI -0.19 to -0.065, p = 4e-04), but not Aβ42 (p = 0.061) or NfL (p = 0.32). Over time, patients with PD had greater increases in serum NfL than controls (β = 0.035, 95% CI 0.022 to 0.048, p = 9.8e-07); slopes of patients with PD did not differ from those of controls for CSF Aβ42 (p = 0.18), p-tau181 (p = 1), or t-tau (p = 0.96). Using ATNPD, PD classified as A+T+N± (n = 32; 9%) had worse cognitive decline on global MoCA (β = -73, 95% CI -110 to -37, p = 0.00077) than all other ATNPD statuses including A+ alone (A+T-N-; n = 75; 21%). DISCUSSION In patients with early PD, CSF p-tau181 and t-tau were low compared with those in controls and did not increase over 5 years of follow-up. Our study shows that classification using modified ATNPD (incorporating CSF Aβ42, CSF p-tau181, and serum NfL) can identify biologically relevant subgroups of PD to improve prediction of cognitive decline in early PD.
Collapse
Affiliation(s)
- Katheryn A Q Cousins
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - David J Irwin
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Thomas F Tropea
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Emma Rhodes
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Jeffrey Phillips
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Alice S Chen-Plotkin
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Michael C Brumm
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Christopher S Coffey
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Ju Hee Kang
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Tanya Simuni
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Tatiana M Foroud
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Arthur W Toga
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Caroline M Tanner
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Karl D Kieburtz
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Brit Mollenhauer
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Douglas Galasko
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Samantha Hutten
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Daniel Weintraub
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Andrew D Siderowf
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Kenneth Marek
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Kathleen L Poston
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| | - Leslie M Shaw
- From the Department of Neurology (K.A.Q.C., D.J.I., T.F.T., E.R., J.P., A.S.C.-P., D.W.), University of Pennsylvania, Philadelphia; Department of Biostatistics (M.C.B., C.S.C.), College of Public Health, University of Iowa, Iowa City; Department of Pharmacology and Clinical Pharmacology (J.H.K.), Inha University, Incheon, South Korea; Feinberg School of Medicine (T.S.), Northwestern University, Chicago, IL; Department of Medical and Molecular Genetics (T.M.F.), Indiana University, Indianapolis; Laboratory of Neuro Imaging (A.W.T.), University of Southern California, Los Angeles; Department of Neurology (C.M.T.), Weill Institute for Neurosciences, University of California San Francisco; Department of Neurology (K.D.K.), University of Rochester Medical Center, NY; Department of Neurology (B.M.), University Medical Center, Göttingen, Paracelsus-Elena-Klinik, Germany; Department of Neurology (D.G.), University of California San Diego; The Michael J. Fox Foundation (S.H.), New York, NY; Department of Psychiatry (D.W.), School of Medicine at the University of Pennsylvania; Michael J. Crescenz VA Medical Center (D.W.), Parkinson's Disease Research, Education, and Clinical Center; Department of Neurology (A.D.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute for Neurodegenerative Disorders (K.M.), New Haven, CT; Department of Neurology (K.L.P.), Stanford University, Palo Alto, CA; and Department of Pathology and Laboratory Medicine (L.M.S.), University of Pennsylvania, Philadelphia
| |
Collapse
|
42
|
Frank A, Bendig J, Schnalke N, Klingelhoefer L, Reichmann H, Akgün K, Ziemssen T, Falkenburger BH. Serum neurofilament indicates accelerated neurodegeneration and predicts mortality in late-stage Parkinson's disease. NPJ Parkinsons Dis 2024; 10:14. [PMID: 38195715 PMCID: PMC10776839 DOI: 10.1038/s41531-023-00605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/17/2023] [Indexed: 01/11/2024] Open
Abstract
Different stages of Parkinson's disease (PD) are defined by clinical criteria, while late-stage PD is marked by the onset of morbidity milestones and rapid clinical deterioration. Based on neuropathological evidence, degeneration in the dopaminergic system occurs primarily in the early stage of PD, raising the question of what drives disease progression in late-stage PD. This study aimed to investigate whether late-stage PD is associated with increased neurodegeneration dynamics rather than functional decompensation using the blood-based biomarker serum neurofilament light chain (sNfL) as a proxy for the rate of neurodegeneration. The study included 118 patients with PD in the transition and late-stage (minimum disease duration 5 years, mean (SD) disease duration 15 (±7) years). The presence of clinical milestones (hallucinations, dementia, recurrent falls, and admission to a nursing home) and mortality were determined based on chart review. We found that sNfL was higher in patients who presented with at least one clinical milestone and increased with a higher number of milestones (Spearman's ρ = 0.66, p < 0.001). Above a cutoff value of 26.9 pg/ml, death was 13.6 times more likely during the follow-up period (95% CI: 3.53-52.3, p < 0.001), corresponding to a sensitivity of 85.0% and a specificity of 85.7% (AUC 0.91, 95% CI: 0.85-0.97). Similar values were obtained when using an age-adjusted cutoff percentile of 90% for sNfL. Our findings suggest that the rate of ongoing neurodegeneration is higher in advanced PD (as defined by the presence of morbidity milestones) than in earlier disease stages. A better understanding of the biological basis of stage-dependent neurodegeneration may facilitate the development of neuroprotective means.
Collapse
Affiliation(s)
- Anika Frank
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.
| | - Jonas Bendig
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nils Schnalke
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lisa Klingelhoefer
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Heinz Reichmann
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Katja Akgün
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Björn H Falkenburger
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| |
Collapse
|
43
|
Chopra A, Outeiro TF. Aggregation and beyond: alpha-synuclein-based biomarkers in synucleinopathies. Brain 2024; 147:81-90. [PMID: 37526295 DOI: 10.1093/brain/awad260] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023] Open
Abstract
Parkinson's disease is clinically known for the loss of dopaminergic neurons in the substantia nigra pars compacta and accumulation of intraneuronal cytoplasmic inclusions rich in alpha-synuclein called 'Lewy bodies' and 'Lewy neurites'. Together with dementia with Lewy bodies and multiple system atrophy, Parkinson's disease is part of a group of disorders called synucleinopathies. Currently, diagnosis of synucleinopathies is based on the clinical assessment which often takes place in advanced disease stages. While the causal role of alpha-synuclein aggregates in these disorders is still debatable, measuring the levels, types or seeding properties of different alpha-synuclein species hold great promise as biomarkers. Recent studies indicate significant differences in peptide, protein and RNA levels in blood samples from patients with Parkinson's disease. Seed amplification assays using CSF, blood, skin biopsy, olfactory swab samples show great promise for detecting synucleinopathies and even for discriminating between different synucleinopathies. Interestingly, small extracellular vesicles, such as exosomes, display differences in their cargoes in Parkinson's disease patients versus controls. In this update, we focus on alpha-synuclein aggregation and possible sources of disease-related species released in extracellular vesicles, which promise to revolutionize the diagnosis and the monitoring of disease progression.
Collapse
Affiliation(s)
- Avika Chopra
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| |
Collapse
|
44
|
Schaeffer E, Kluge A, Schulte C, Deuschle C, Bunk J, Welzel J, Maetzler W, Berg D. Association of Misfolded α-Synuclein Derived from Neuronal Exosomes in Blood with Parkinson's Disease Diagnosis and Duration. JOURNAL OF PARKINSON'S DISEASE 2024; 14:667-679. [PMID: 38669557 PMCID: PMC11191501 DOI: 10.3233/jpd-230390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 04/28/2024]
Abstract
Background Misfolded α-synuclein can be detected in blood samples of Parkinson's disease (PD) patients by a seed amplification assay (SAA), but the association with disease duration is not clear, yet. Objective In the present study we aimed to elucidate whether seeding activity of misfolded α-synuclein derived from neuronal exosomes in blood is associated with PD diagnosis and disease duration. Methods Cross-sectional samples of PD patients were analyzed and compared to samples of age- and gender-matched healthy controls using a blood-based SAA. Presence of α-synuclein seeding activity and differences in seeding parameters, including fluorescence response (in arbitrary units) at the end of the amplification assay (F60) were analyzed. Additionally, available PD samples collected longitudinally over 5-9 years were included. Results In the cross-sectional dataset, 79 of 80 PD patients (mean age 69 years, SD = 8; 56% male) and none of the healthy controls (n = 20, mean age 70 years, SD = 10; 55% male) showed seeding activity (sensitivity 98.8%). When comparing subgroups divided by disease duration, longer disease duration was associated with lower α-synuclein seeding activity (F60: p < 0.001). In the longitudinal analysis 10/11 patients showed a gradual decrease of α-synuclein seeding activity over time. Conclusions This study confirms the high sensitivity of the blood-based α-synuclein SAA applied here. The negative association of α-synuclein seeding activity in blood with disease duration makes this parameter potentially interesting as biomarker for future studies on the pathophysiology of disease progression in PD, and for biologically oriented trials in this field.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Annika Kluge
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Christian Deuschle
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Josina Bunk
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Julius Welzel
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| |
Collapse
|
45
|
Starmans NLP, Kappelle LJ, Muller M, Staals J, Teunissen CE, Biessels GJ, van der Flier WM, Wolters FJ. Blood Pressure Variability and Plasma Biomarkers of Neuronal Injury and Alzheimer's Disease: A Clinic-Based Study of Patients with Diseases Along the Heart-Brain Axis. J Alzheimers Dis 2024; 99:1207-1215. [PMID: 38788076 PMCID: PMC11191465 DOI: 10.3233/jad-240119] [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] [Accepted: 04/10/2024] [Indexed: 05/26/2024]
Abstract
Higher blood pressure variability (BPV) predisposes to cognitive decline. To investigate underlying mechanisms, we measured 24-h ambulatory BPV, nocturnal dipping and orthostatic hypotension in 518 participants with vascular cognitive impairment, carotid occlusive disease, heart failure, or reference participants. We determined cross-sectional associations between BPV indices and plasma biomarkers of neuronal injury (neurofilament light chain) and Alzheimer's disease (phosphorylated-tau-181 and Aβ42/Aβ40). None of the BPV indices were significantly associated with any of the biomarkers. Hence, in patients with diseases along the heart-brain axis, we found no evidence for an association between BPV and selected markers of neuronal injury or Alzheimer's disease.
Collapse
Affiliation(s)
| | - Laurens Jaap Kappelle
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Majon Muller
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Julie Staals
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Charlotte Elisabeth Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiesje Maria van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank Johannes Wolters
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - on behalf of the Heart-Brain Connection Consortium
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Internal Medicine, Geriatrics Section, Amsterdam Cardiovascular Science, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Department of Epidemiology, Amsterdam University Medical Center (Amsterdam UMC), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine and Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
46
|
Xiang Y, Huang X, Xu Q, Liu Z, Chen Y, Sun Q, Wang J, Jiang H, Shen L, Yan X, Tang B, Guo J. Estimating the sequence of biomarker changes in Parkinson's disease. Parkinsonism Relat Disord 2024; 118:105939. [PMID: 38029648 DOI: 10.1016/j.parkreldis.2023.105939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/30/2023] [Accepted: 11/19/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE To estimate the sequence of several common biomarker changes in Parkinson's disease (PD) using a novel data-driven method. METHODS We included 374 PD patients and 169 healthy controls (HC) from the Parkinson's Progression Markers Initiative (PPMI). Biomarkers, including the left putamen striatal binding ratio (SBR), right putamen SBR, left caudate SBR, right caudate SBR, cerebrospinal fluid (CSF) α-synuclein, and serum neurofilament light chain (NfL), were selected in our study. The discriminative event-based model (DEBM) was utilized to model the sequence of biomarker changes and establish the disease progression timeline. The estimated disease stages for each subject were obtained through cross-validation. The associations between the estimated disease stages and the clinical symptoms of PD were explored using Spearman's correlation. RESULTS The left putamen is the earliest biomarker to become abnormal among the selected biomarkers, followed by the right putamen, CSF α-synuclein, right caudate, left caudate, and serum NfL. The estimated disease stages are significantly different between PD and HC and yield a high accuracy for distinguishing PD from HC, with an area under the curve (AUC) of 0.98 (95% confidence interval 0.97-0.99), a sensitivity of 0.95, and a specificity of 0.92. Moreover, the estimated disease stages correlate with motor experiences of daily living, motor symptoms, autonomic dysfunction, and anxiety in PD patients. CONCLUSION We determined the sequence of several common biomarker changes in PD using DEBM, providing data-driven evidence of the disease progression of PD.
Collapse
Affiliation(s)
- Yaqin Xiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - XiuRong Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenhua Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yase Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiying Sun
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Centre for Medical Genetics, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Centre for Medical Genetics, Central South University, Changsha, China.
| |
Collapse
|
47
|
Gątarek P, Kałużna-Czaplińska J. Integrated metabolomics and proteomics analysis of plasma lipid metabolism in Parkinson's disease. Expert Rev Proteomics 2024; 21:13-25. [PMID: 38346207 DOI: 10.1080/14789450.2024.2315193] [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: 11/19/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Metabolomics and proteomics are two growing fields of science which may shed light on the molecular mechanisms that contribute to neurodegenerative diseases. Studies focusing on these aspects can reveal specific metabolites and proteins that can halt or reverse the progressive neurodegenerative process leading to dopaminergic cell death in the brain. AREAS COVERED In this article, an overview of the current status of metabolomic and proteomic profiling in the neurodegenerative disease such as Parkinson's disease (PD) is presented. We discuss the importance of state-of-the-art metabolomics and proteomics using advanced analytical methodologies and their potential for discovering new biomarkers in PD. We critically review the research to date, highlighting how metabolomics and proteomics can have an important impact on early disease diagnosis, future therapy development and the identification of new biomarkers. Finally, we will discuss interactions between lipids and α-synuclein (SNCA) and also consider the role of SNCA in lipid metabolism. EXPERT OPINION Metabolomic and proteomic studies contribute to understanding the biological basis of PD pathogenesis, identifying potential biomarkers and introducing new therapeutic strategies. The complexity and multifactorial nature of this disease requires a comprehensive approach, which can be achieved by integrating just these two omic studies.
Collapse
Affiliation(s)
- Paulina Gątarek
- Institute Of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| | - Joanna Kałużna-Czaplińska
- Institute Of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
48
|
Liu X, Chen J, Meng C, Zhou L, Liu Y. Serum neurofilament light chain and cognition decline in US elderly: A cross-sectional study. Ann Clin Transl Neurol 2024; 11:17-29. [PMID: 37902309 PMCID: PMC10791034 DOI: 10.1002/acn3.51929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
OBJECTIVE Early identification of cognitive impairment in neurodegenerative diseases like Alzheimer's disease (AD) is crucial. Neurofilament, a potential biomarker for neurological disorders, has gained attention. Our study aims to investigate the relationship between serum neurofilament light (sNfL) levels and cognitive function in elderly individuals in the United States. METHODS This cross-sectional study analyzed data from participants aged 60 and above in the National Health and Nutrition Examination Survey (2013-2014). We collected sNfL levels, cognitive function tests, sociodemographic characteristics, comorbidities, and other variables. Weighted multiple linear regression models examined the relationship between ln(sNfL) and cognitive scores. Restricted cubic spline (RCS) visualization explored nonlinear relationships. The stratified analysis examined subgroups' ln(sNfL) and cognitive function association. RESULTS The study included 446 participants (47.73% male). Participants with ln(sNfL) levels between 2.58 and 2.81 pg/mL (second quintile) performed relatively well in cognitive tests. After adjusting for multiple factors, ln(sNfL) levels were negatively correlated with cognitive function, with adjusted β (95% CI) as follows: immediate recall test (IRT): -0.763 (-1.301 to -0.224), delayed recall test (DRT): -0.308 (-0.576 to -0.04), animal fluency test (AFT): -1.616 (-2.639 to -0.594), and digit symbol substitution test (DSST): -2.790 (-4.369 to -1.21). RCS curves showed nonlinear relationships between ln(sNfL) and DRT, AFT, with inflection points around 2.7 pg/mL. The stratified analysis revealed a negative correlation between ln(sNfL) and cognition in specific subgroups with distinct features, with an interaction between diabetes and ln(sNfL). INTERPRETATION Higher sNfL levels are associated with poorer cognitive function in the elderly population of the United States. sNfL shows promise as a potential biomarker for early identification of cognitive decline.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Jun Chen
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Chen Meng
- Department of Anesthesiology, Taihe HospitalHubei University of MedicineShiyanHubeiChina
| | - Lan Zhou
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| | - Yong Liu
- Department of Neurology, Taihe HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
49
|
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
|
50
|
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
|