1
|
Mao-Draayer Y, Bar-Or A, Balashov K, Foley J, Smoot K, Longbrake EE, Robertson D, Mendoza JP, Lewin JB, Everage N, Božin I, Lyons J, Mokliatchouk O, Bame E, Giuliani F. Real-World Safety and Effectiveness of Dimethyl Fumarate in Patients with MS: Results from the ESTEEM Phase 4 and PROCLAIM Phase 3 Studies with a Focus on Older Patients. Adv Ther 2025; 42:395-412. [PMID: 39570545 PMCID: PMC11782338 DOI: 10.1007/s12325-024-03047-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
INTRODUCTION Real-world studies in the USA report that 41-56% of patients with multiple sclerosis (MS) are ≥ 50 years old, yet data on their response to disease-modifying therapies (DMTs) is limited. Dimethyl fumarate (DMF) is an oral DMT approved for treating relapsing MS. This analysis evaluated the safety, efficacy, and immunophenotype changes of DMF in patients ≥ 50 years compared with patients < 50 years. METHODS ESTEEM, a 5-year, real-world, observational phase 4 study, assessed the safety and effectiveness of DMF, including treatment-emergent serious adverse events (SAEs) and adverse events (AEs) leading to treatment discontinuation. Absolute lymphocyte counts (ALCs) were recorded from a subset of patients. The PROCLAIM study, a phase 3b interventional study, reported safety outcomes and lymphocyte subset changes in patients with relapsing-remitting MS (RRMS) treated with DMF. The study evaluated safety outcomes by analyzing the incidence of SAEs and detailed changes in CD4+ and CD8+ T cell compartments over 96 weeks of DMF treatment. RESULTS ESTEEM included 4020 patients aged < 50 years and 1069 aged ≥ 50 years. AEs leading to discontinuation were reported by 19.6% patients < 50 years and 29.6% of patients ≥ 50 years, with gastrointestinal disorders being the most common. SAEs were reported by 5.2% of patients < 50 years and 8.9% those ≥ 50 years. In PROCLAIM, SAEs were reported in 13% of patients < 50 years and 10% of those ≥ 50 years. Median ALC decreased by 35% in patients < 50 years and 50% in those ≥ 50 years in ESTEEM, with similar patterns observed in PROCLAIM. CONCLUSIONS ESTEEM found no unexpected safety signals in older patients and annualized relapse rates (ARRs) were significantly reduced in both age groups. Both studies indicated that DMF is efficacious and has a favorable safety profile in patients with RRMS aged ≥ 50 years. CLINICAL TRIAL REGISTRATION ESTEEM (NCT02047097), PROCLAIM (NCT02525874).
Collapse
Affiliation(s)
| | - Amit Bar-Or
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - John Foley
- Rocky Mountain Multiple Sclerosis Clinic, Salt Lake City, UT, USA
| | - Kyle Smoot
- Providence MS Center, Providence Brain and Spine Institute, Portland, OR, USA
| | | | | | | | | | | | | | | | | | - Eris Bame
- Biogen, Cambridge, MA, USA
- Department of Internal Medicine and Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | |
Collapse
|
2
|
Trifilio E, Bottari S, McQuillan LE, Barton DJ, Lamb DG, Robertson C, Rubenstein R, Wang KK, Wagner AK, Williamson JB. Temporal Profile of Serum Neurofilament Light (NF-L) and Heavy (pNF-H) Level Associations With 6-Month Cognitive Performance in Patients With Moderate-Severe Traumatic Brain Injury. J Head Trauma Rehabil 2024; 39:E470-E480. [PMID: 38758056 PMCID: PMC11534502 DOI: 10.1097/htr.0000000000000932] [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] [Indexed: 05/18/2024]
Abstract
OBJECTIVE Identification of biomarkers of cognitive recovery after traumatic brain injury (TBI) will inform care and improve outcomes. This study assessed the utility of neurofilament (NF-L and pNF-H), a marker of neuronal injury, informing cognitive performance following moderate-to-severe TBI (msTBI). SETTING Level 1 trauma center and outpatient via postdischarge follow-up. PARTICIPANTS N = 94. Inclusion criteria : Glasgow Coma Scale score less than 13 or 13-15 with clinical evidence of moderate-to-severe injury traumatic brain injury on clinical imaging. Exclusion criteria : neurodegenerative condition, brain death within 3 days after injury. DESIGN Prospective observational study. Blood samples were collected at several time points post-injury. Cognitive testing was completed at 6 months post-injury. MAIN MEASURES Serum NF-L (Human Neurology 4-Plex B) pNF-H (SR-X) as measured by SIMOA Quanterix assay. Divided into 3 categorical time points at days post-injury (DPI): 0-15 DPI, 16-90 DPI, and >90 DPI. Cognitive composite comprised executive functioning measures derived from 3 standardized neuropsychological tests (eg, Delis-Kaplan Executive Function System: Verbal Fluency, California Verbal Learning Test, Second Edition, Wechsler Adult Intelligence Scale, Third Edition). RESULTS pNF-H at 16-90 DPI was associated with cognitive outcomes including a cognitive-executive composite score at 6 months ( β = -.430, t34 = -3.190, P = .003). CONCLUSIONS Results suggest that "subacute" elevation of serum pNF-H levels may be associated with protracted/poor cognitive recovery from msTBI and may be a target for intervention. Interpretation is limited by small sample size and including only those who were able to complete cognitive testing.
Collapse
Affiliation(s)
- Erin Trifilio
- Author Affiliations: Brain Rehabilitation Research Center (BRRC), Malcom Randall VAMC, Gainesville, Florida (Drs Trifilio, Lamb, Wang, and Williamson and Ms Bottari); Department of Clinical and Health Psychology (Drs Trifilio and Williamson and Ms Bottari), College of Public Health and Health Professions, and Departments of Emergency Medicine (Dr Wang) and Psychiatry (Drs Lamb and Williamson), College of Medicine, University of Florida, Gainesville; Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (Dr Robertson); Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York (Dr Rubenstein); Department of Physical Medicine and Rehabilitation (Ms McQuillan and Dr Wagner), Department of Emergency Medicine (Dr Barton), Department of Neuroscience (Dr Wagner), Clinical and Translational Science Institute (Dr Wagner), and Safar Center for Resuscitation Research (Dr Wagner); University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia (Dr Wang)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Tisell A, Söderberg K, Link Y, Lundberg P, Mellergård J. Diffuse white matter pathology in multiple sclerosis during treatment with dimethyl fumarate-An observational study of changes in normal-appearing white matter using proton magnetic resonance spectroscopy. PLoS One 2024; 19:e0309547. [PMID: 39432495 PMCID: PMC11493296 DOI: 10.1371/journal.pone.0309547] [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: 09/20/2023] [Accepted: 08/13/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory demyelinating disease with neurodegenerative features causing risk for neurologic irreversible disability over time. Examination of normal-appearing white matter (NAWM) changes in MS by proton magnetic resonance spectroscopy (1H-MRS), may detect diffuse white matter pathology that is associated with neurodegeneration. METHODS In this observational study of in total twenty-six patients with MS, starting treatment with dimethyl fumarate (DMF), we measured the absolute concentration of metabolites in periventricular NAWM using 1H-MRS at baseline and after one and three years of treatment. Metabolite concentrations were analyzed both cross-sectionally, in relation to 10 controls and longitudinally in relation to disease activity. RESULTS Patients with MS had higher concentrations of myo-inositol (mIns) in NAWM at baseline compared with controls (mean 5.98 ± 1.37 (SD) and 4.32 ± 1.16 (SD), p<0.01, independent samples t-test). The disease duration was inversely correlated with concentrations of total N-acetylaspartate and N-acetylaspartylglutamate (tNA) (r = -0.62, p<0.01) in NAWM as well as positively to the ratio of mIns and tNA (r = 0.51, p = 0.03). Metabolite concentrations during one-year (n = 19) and three-years (n = 11) follow-up were generally stable. The dropouts were caused by treatment switch after one year, mainly due to new MRI activity. Cross-sectional analyses showed that there was an inverse correlation between concentrations of tNA and mIns at both baseline and at 1 and 3-years follow-up (r = -0.44 to -0.65, p = 0.04 to 0.004). Metabolite concentrations were stable during 1-year follow-up independently of disease activity. CONCLUSIONS Higher concentrations of the astrogliosis marker mIns in MS compared to controls, the inverse relation between MS disease duration and the neuroaxonal integrity marker tNA, as well as the consistent inverse relation between these two metabolites during follow-up, showed that non-lesional white matter pathology is present in this cohort of MS patients in early disease stages. However, metabolite concentrations during follow-up were generally stable and did not reflect differences in disease activity among patients.
Collapse
Affiliation(s)
- Anders Tisell
- Department of Medical Radiation Physics in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Kristina Söderberg
- Department of Radiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Yumin Link
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Department of Medical Radiation Physics in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Johan Mellergård
- Department of Neurology in Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Rosas HD, Mercaldo ND, Hasimoglu Y, Petersen M, Lewis LR, Lai F, Powell D, Dhungana A, Demir A, Keater D, Yassa M, Brickman AM, O'Bryant S. Association of plasma neurofilament light chain with microstructural white matter changes in Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70023. [PMID: 39583646 PMCID: PMC11582681 DOI: 10.1002/dad2.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Both micro- and macrostructural white matter (WM) abnormalities, particularly those related to axonal degeneration, are associated with cognitive decline in adults with Down syndrome (DS) prior to a diagnosis of Alzheimer disease. Neurofilament light chain (NfL) is a support protein within myelinated axons released into blood following axonal damage. In this study we investigated cross-sectional relationships between WM microstructural changes as measured by diffusion tensor imaging (DTI) and plasma NfL concentration in adults with DS without dementia. METHODS Thirty cognitively stable (CS) adults with DS underwent diffusion-weighted MRI scanning and plasma NfL measurement. DTI measures of select WM tracts were derived using automatic fiber tracking, and associations with plasma NfL were assessed using Spearman correlation coefficients. RESULTS Higher Plasma NfL was associated with greater altered diffusion measures of select tracts. DISCUSSION Early increases in plasma NfL may reflect early white matter microstructural changes prior to dementia in DS. Highlights The onset of such WM changes in DS has not yet been widely studied.WM microstructural properties correlated with plasma neurofilament light chain (NfL).NfL may reflect early, selective WM changes in adults with DS at high risk of developing AD.
Collapse
Affiliation(s)
- Herminia Diana Rosas
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Nathaniel David Mercaldo
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Yasemin Hasimoglu
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Melissa Petersen
- Department of Family MedicineUniversity of North Texas Health Science CenterFort WorthTexasUSA
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Lydia R. Lewis
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Florence Lai
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - David Powell
- Magnetic Resonance Imaging and Spectroscopy CenterUniversity of KentuckyLexingtonKentuckyUSA
| | - Asim Dhungana
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Ali Demir
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Neuroimaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalCharlestownMassachusettsUSA
| | - David Keater
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Michael Yassa
- Department of Psychiatry and Human BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's disease and the Aging Brain Department of NeurologyVagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUSA
| | - Sid O'Bryant
- Department of Family MedicineUniversity of North Texas Health Science CenterFort WorthTexasUSA
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
5
|
Bava CI, Valentino P, Malucchi S, Bottero R, Martire S, Sapio AD, Bertolotto A. Prevalence of elevated sNFL in a real-world setting: Results on 908 patients with different multiple sclerosis types and treatment conditions. Mult Scler Relat Disord 2024; 88:105748. [PMID: 38959590 DOI: 10.1016/j.msard.2024.105748] [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: 12/29/2023] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND In the field of research for new validated surrogate biomarkers of treatment efficacy, disease activity and progression in Multiple Sclerosis (MS), serum neurofilament light-chain (sNFL) are actually the best candidate for MS patient monitoring. However, before they can be implemented in clinical practice, their usefulness as additional red flag routine measure must be demonstrated. To tackle the problem, this real-life cross-sectional study at the Regional Referring Center for Multiple Sclerosis (CRESM) aims to characterize sNFL levels and prevalence of elevated sNFL, according to our age-dependent cut-off values, in a large group of patients with different types of MS and treatment conditions. METHODS 908 serum samples from as many MS patients being admitted at CRESM for diagnostic definition and/or during routinary treatment monitoring were consecutively collected between January 2019 and January 2020. sNFL levels were measured by single molecule array (Simoa™) technology on SR-X instrument using NF-light assays (Quanterix); results were interpreted using previously published cut-off values. RESULTS Primary and Secondary Progressive MS (PPMS, SPMS) forms demonstrate higher levels and prevalence of elevated sNFL (PPMS= 32 %, SPMS= 21 %) compared to the Relapse and Remitting one (RRMS = 12 %). Besides, naïve samples of RRMS and PPMS subtypes showed higher prevalence of elevated sNFL (RRMS naïve= 31 %, PPMS naïve=67 %) compared to samples from patients treated for more than 12 months (RRMS treat>12m= 9 %, PPMS treat>12m= 19 %); treated SPMS patients demonstrated higher sNFL levels and a prevalence (22 %) of elevated sNFL compared to RRMS treated patients. Focusing on RRMS, no statistical difference was found between groups of patients treated for whatever time (up to or more than 60 months) and with either DMT type (high or low-efficacy DMT). Finally, RRMS patients treated with all DMTs for more than 12 months, with the exception of teriflunomide and alemtuzumab showed a prevalence of elevated sNFL in the range of 5-10 %. CONCLUSION in a real-world setting comprising about 1000 MS patients, sNFL quantification was elevated in 5-to-67 % of patients, in different MS forms and treatment conditions. Elevated levels of sNFL must be considered a red-flag suggesting the need of a further clinical monitoring in any circumstance, as it can be indicative of new inflammation, ongoing degeneration or co-morbidities. This study supports the introduction of sNFL quantification in everyday patient management.
Collapse
Affiliation(s)
- Cecilia Irene Bava
- NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy; CRESM Biobank, University Hospital San Luigi Gonzaga, Orbassano, Italy; Department of Neurology and CRESM, University Hospital San Luigi Gonzaga, Orbassano, Italy.
| | - Paola Valentino
- NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Simona Malucchi
- Department of Neurology and CRESM, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | - Rugiada Bottero
- Department of Neurology and CRESM, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | - Serena Martire
- CRESM Biobank, University Hospital San Luigi Gonzaga, Orbassano, Italy; Department of Neuroscience "Rita Levi Montalcini", University of Turin, Italy
| | - Alessia Di Sapio
- CRESM Biobank, University Hospital San Luigi Gonzaga, Orbassano, Italy; Department of Neurology and CRESM, University Hospital San Luigi Gonzaga, Orbassano, Italy
| | | |
Collapse
|
6
|
Sung J, Chae Y, Yun T, Koo Y, Lee D, Kim H, Yang M, Kang B. Use of neurofilament light chain to identify structural brain diseases in dogs. J Vet Intern Med 2024; 38:2196-2203. [PMID: 38778568 PMCID: PMC11256173 DOI: 10.1111/jvim.17110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Neurofilament light chain (NfL) is released into the peripheral circulation by damaged axons. OBJECTIVES To evaluate the diagnostic value of serum NfL concentration in dogs with intracranial diseases. ANIMALS Study included 37 healthy dogs, 31 dogs with idiopathic epilepsy (IE), 45 dogs with meningoencephalitis of unknown etiology (MUE), 20 dogs with hydrocephalus, and 19 dogs with brain tumors. METHODS Cohort study. Serum NfL concentrations were measured in all dogs using single-molecule array technology. RESULTS Serum NfL concentration in dogs with each structural disease was significantly higher than in healthy dogs and dogs with IE (P = .01). The area under the receiver operating characteristic curve of NfL for differentiating between dogs with structural diseases and IE was 0.868. An optimal cutoff value of the NfL 27.10 pg/mL had a sensitivity of 86.67% and a specificity of 74.19% to differentiate the dogs with IE from those with structural brain diseases. There were significant correlations between NfL concentrations and lesion size: (1) MUE, P = .01, r = 0.429; (2) hydrocephalus, P = .01, r = 0.563. CONCLUSIONS AND CLINICAL IMPORTANCE Serum NfL could be a useful biomarker for distinguishing IE from structural diseases in dogs and predicting the lesion sizes of MUE and hydrocephalus.
Collapse
Affiliation(s)
- Jookyung Sung
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Yeon Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Taesik Yun
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Yoonhoi Koo
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Dohee Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Hakhyun Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Mhan‐Pyo Yang
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Byeong‐Teck Kang
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| |
Collapse
|
7
|
Schaller-Paule MA, Maiworm M, Schäfer JH, Friedauer L, Hattingen E, Wenger KJ, Weber F, Jakob J, Steffen F, Bittner S, Yalachkov Y, Foerch C. Matching proposed clinical and MRI criteria of aggressive multiple sclerosis to serum and cerebrospinal fluid markers of neuroaxonal and glial injury. J Neurol 2024; 271:3512-3526. [PMID: 38536455 PMCID: PMC11136815 DOI: 10.1007/s00415-024-12299-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Definitions of aggressive MS employ clinical and MR imaging criteria to identify highly active, rapidly progressing disease courses. However, the degree of overlap between clinical and radiological parameters and biochemical markers of CNS injury is not fully understood. Aim of this cross-sectional study was to match clinical and MR imaging hallmarks of aggressive MS to serum/CSF markers of neuroaxonal and astroglial injury (neurofilament light chain (sNfL, cNfL), and glial fibrillary acidic protein (sGFAP, cGFAP)). METHODS We recruited 77 patients with relapsing-remitting MS (RRMS) and 22 patients with clinically isolated syndrome. NfL and GFAP levels in serum and CSF were assessed using a single-molecule-array HD-1-analyzer. A general linear model with each biomarker as a dependent variable was computed. Clinical and imaging criteria of aggressive MS, as recently proposed by the ECTRIMS Consensus Group, were modeled as independent variables. Other demographic, clinical or laboratory parameters, were modeled as covariates. Analyses were repeated in a homogenous subgroup, consisting only of newly diagnosed, treatment-naïve RRMS patients presenting with an acute relapse. RESULTS After adjusting for covariates and multiplicity of testing, sNfL and cNfL concentrations were strongly associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.00008; pcNfL = 0.004) as well as the presence of infratentorial lesions on MRI (psNfL = 0.0003; pcNfL < 0.004). No other clinical and imaging criteria of aggressive MS correlated significantly with NfL or GFAP in serum and CSF. In the more homogeneous subgroup, sNfL still was associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.001), presence of more than 20 T2-lesions (psNfL = 0.049) as well as the presence of infratentorial lesions on MRI (psNfL = 0.034), while cNfL was associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.011) and presence of more than 20 T2-lesions (psNfL = 0.029). CONCLUSIONS Among proposed risk factors for an aggressive disease course, MRI findings but not clinical characteristics correlated with sNfL and cNfL as a marker of neuroaxonal injury and should be given appropriate weight considering MS prognosis and therapy. No significant correlation was detected for GFAP alone.
Collapse
Affiliation(s)
- Martin A Schaller-Paule
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany.
- Practice for Neurology and Psychiatry Eltville, 65343, Eltville, Germany.
| | - Michelle Maiworm
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
| | - Jan Hendrik Schäfer
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
| | - Lucie Friedauer
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Katharina Johanna Wenger
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Jasmin Jakob
- 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
| | - Falk Steffen
- 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
| | - 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
| | - Yavor Yalachkov
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
| | - Christian Foerch
- Department of Neurology, University Hospital Frankfurt, Goethe University Frankfurt, Schleusenweg 2-16, 60528, Frankfurt, Germany
| |
Collapse
|
8
|
Tato-Fernández C, Ekblad LL, Pietilä E, Saunavaara V, Helin S, Parkkola R, Zetterberg H, Blennow K, Rinne JO, Snellman A. Cognitively healthy APOE4/4 carriers show white matter impairment associated with serum NfL and amyloid-PET. Neurobiol Dis 2024; 192:106439. [PMID: 38365046 DOI: 10.1016/j.nbd.2024.106439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024] Open
Abstract
Except for aging, carrying the APOE ε4 allele (APOE4) is the most important risk factor for sporadic Alzheimer's disease. APOE4 carriers may have reduced capacity to recycle lipids, resulting in white matter microstructural abnormalities. In this study, we evaluated whether white matter impairment measured by diffusion tensor imaging (DTI) differs between healthy individuals with a different number of APOE4 alleles, and whether white matter impairment associates with brain beta-amyloid (Aβ) load and serum levels of neurofilament light chain (NfL). We studied 96 participants (APOE3/3, N = 37; APOE3/4, N = 39; APOE4/4, N = 20; mean age 70.7 (SD 5.22) years, 63% females) with a brain MRI including a DTI sequence (N = 96), Aβ-PET (N = 89) and a venous blood sample for the serum NfL concentration measurement (N = 88). Fractional anisotropy (FA), mean diffusivity (MD), radial diffusivity (RD) and axial diffusivity (AxD) in six a priori-selected white matter regions-of-interest (ROIs) were compared between the groups using ANCOVA, with sex and age as covariates. A voxel-weighted average of FA, MD, RD and AxD was calculated for each subject, and correlations with Aβ-PET and NfL levels were evaluated. APOE4/4 carriers exhibited a higher MD and a higher RD in the body of corpus callosum than APOE3/4 (p = 0.0053 and p = 0.0049, respectively) and APOE3/3 (p = 0.026 and p = 0.042). APOE4/4 carriers had a higher AxD than APOE3/4 (p = 0.012) and APOE3/3 (p = 0.040) in the right cingulum adjacent to cingulate cortex. In the total sample, composite MD, RD and AxD positively correlated with the cortical Aβ load (r = 0.26 to 0.33, p < 0.013 for all) and with serum NfL concentrations (r = 0.31 to 0.36, p < 0.0028 for all). In conclusion, increased local diffusivity was detected in cognitively unimpaired APOE4/4 homozygotes compared to APOE3/4 and APOE3/3 carriers, and increased diffusivity correlated with biomarkers of Alzheimer's disease and neurodegeneration. White matter impairment seems to be an early phenomenon in the Alzheimer's disease pathologic process in APOE4/4 homozygotes.
Collapse
Affiliation(s)
- Claudia Tato-Fernández
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland.
| | - Laura L Ekblad
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Geriatric Medicine, Turku University Hospital, Turku, Finland
| | - Elina Pietilä
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland
| | - Virva Saunavaara
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Medical Physics, Division of Medical Imaging, Turku University Hospital, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, Turku, Finland; Department of Radiology, University of Turku, Turku, Finland
| | - 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
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Juha O Rinne
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Anniina Snellman
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland
| |
Collapse
|
9
|
Hermesdorf M, Wulms N, Maceski A, Leppert D, Benkert P, Wiendl H, Kuhle J, Berger K. Serum neurofilament light and white matter characteristics in the general population: a longitudinal analysis. GeroScience 2024; 46:463-472. [PMID: 37285009 PMCID: PMC10828306 DOI: 10.1007/s11357-023-00846-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/24/2023] [Indexed: 06/08/2023] Open
Abstract
Neurofilament light polypeptide (NfL) is a component of the neuronal cytoskeleton and particularly abundant in large-caliber axons. When axonal injury occurs, NfL is released and reaches the cerebrospinal fluid and the blood. Associations between NfL and white matter alterations have previously been observed in studies based on patients with neurological diseases. The current study aimed to explore the relationship between serum NfL (sNfL) and white matter characteristics in a population-based sample. The cross-sectional associations between sNfL as dependent variable, fractional anisotropy (FA), and white matter lesion (WML) volume were analyzed with linear regression models in 307 community-dwelling adults aged between 35 and 65 years. These analyses were repeated with additional adjustment for the potential confounders age, sex, and body mass index (BMI). Longitudinal associations over a mean follow-up of 5.39 years were analyzed with linear mixed models. The unadjusted cross-sectional models yielded significant associations between sNfL, WML volume, and FA, respectively. However, after the adjustment for confounders, these associations did not reach significance. In the longitudinal analyses, the findings corroborated the baseline findings showing no significant associations between sNfL and white matter macrostructure and microstructure beyond the effects of age. In synopsis with previous studies in patients with acute neurological diseases showing a significant association of sNfL with white matter changes beyond the effects of age, the present results based on a sample from the general population suggest the perspective that changes in sNfL reflect age-related effects that also manifest in altered white matter macrostructure and microstructure.
Collapse
Affiliation(s)
- Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany.
| | - Niklas Wulms
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Aleksandra Maceski
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - David Leppert
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Heinz Wiendl
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Jens Kuhle
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| |
Collapse
|
10
|
Chong JSX, Tan YJ, Koh AJ, Ting SKS, Kandiah N, Ng ASL, Zhou JH. Plasma Neurofilament Light Relates to Divergent Default and Salience Network Connectivity in Alzheimer's Disease and Behavioral Variant Frontotemporal Dementia. J Alzheimers Dis 2024; 99:965-980. [PMID: 38759005 PMCID: PMC11191491 DOI: 10.3233/jad-231251] [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: 03/27/2024] [Indexed: 05/19/2024]
Abstract
Background Alzheimer's disease (AD) and behavioral variant frontotemporal dementia (bvFTD) show differential vulnerability to large-scale brain functional networks. Plasma neurofilament light (NfL), a promising biomarker of neurodegeneration, has been linked in AD patients to glucose metabolism changes in AD-related regions. However, it is unknown whether plasma NfL would be similarly associated with disease-specific functional connectivity changes in AD and bvFTD. Objective Our study examined the associations between plasma NfL and functional connectivity of the default mode and salience networks in patients with AD and bvFTD. Methods Plasma NfL and neuroimaging data from patients with bvFTD (n = 16) and AD or mild cognitive impairment (n = 38; AD + MCI) were analyzed. Seed-based functional connectivity maps of key regions within the default mode and salience networks were obtained and associated with plasma NfL in these patients. RESULTS We demonstrated divergent associations between NfL and functional connectivity in AD + MCI and bvFTD patients. Specifically, AD + MCI patients showed lower default mode network functional connectivity with higher plasma NfL, while bvFTD patients showed lower salience network functional connectivity with higher plasma NfL. Further, lower NfL-related default mode network connectivity in AD + MCI patients was associated with lower Montreal Cognitive Assessment scores and higher Clinical Dementia Rating sum-of-boxes scores, although NfL-related salience network connectivity in bvFTD patients was not associated with Neuropsychiatric Inventory Questionnaire scores. CONCLUSIONS Our findings indicate that plasma NfL is differentially associated with brain functional connectivity changes in AD and bvFTD.
Collapse
Affiliation(s)
- Joanna Su Xian Chong
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Human Potential Translational Research Programme and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Amelia Jialing Koh
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Human Potential Translational Research Programme and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Simon Kang Seng Ting
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Adeline Su Lyn Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Human Potential Translational Research Programme and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), NUS Graduate School, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore
| |
Collapse
|
11
|
Elmers J, Colzato LS, Akgün K, Ziemssen T, Beste C. Neurofilaments - Small proteins of physiological significance and predictive power for future neurodegeneration and cognitive decline across the life span. Ageing Res Rev 2023; 90:102037. [PMID: 37619618 DOI: 10.1016/j.arr.2023.102037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/15/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Neurofilaments (NFs) are not only important for axonal integrity and nerve conduction in large myelinated axons but they are also thought to be crucial for receptor and synaptic functioning. Therefore, NFs may play a critical role in cognitive functions, as cognitive processes are known to depend on synaptic integrity and are modulated by dopaminergic signaling. Here, we present a theory-driven interdisciplinary approach that NFs may link inflammation, neurodegeneration, and cognitive functions. We base our hypothesis on a wealth of evidence suggesting a causal link between inflammation and neurodegeneration and between these two and cognitive decline (see Fig. 1), also taking dopaminergic signaling into account. We conclude that NFs may not only serve as biomarkers for inflammatory, neurodegenerative, and cognitive processes but also represent a potential mechanical hinge between them, moreover, they may even have predictive power regarding future cognitive decline. In addition, we advocate the use of both NFs and MRI parameters, as their synthesis offers the opportunity to individualize medical treatment by providing a comprehensive view of underlying disease activity in neurological diseases. Since our society will become significantly older in the upcoming years and decades, maintaining cognitive functions and healthy aging will play an important role. Thanks to technological advances in recent decades, NFs could serve as a rapid, noninvasive, and relatively inexpensive early warning system to identify individuals at increased risk for cognitive decline and could facilitate the management of cognitive dysfunctions across the lifespan.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| | - Katja Akgün
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
12
|
Ellen O, Ye S, Nheu D, Dass M, Pagnin M, Ozturk E, Theotokis P, Grigoriadis N, Petratos S. The Heterogeneous Multiple Sclerosis Lesion: How Can We Assess and Modify a Degenerating Lesion? Int J Mol Sci 2023; 24:11112. [PMID: 37446290 DOI: 10.3390/ijms241311112] [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: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous disease of the central nervous system that is governed by neural tissue loss and dystrophy during its progressive phase, with complex reactive pathological cellular changes. The immune-mediated mechanisms that promulgate the demyelinating lesions during relapses of acute episodes are not characteristic of chronic lesions during progressive MS. This has limited our capacity to target the disease effectively as it evolves within the central nervous system white and gray matter, thereby leaving neurologists without effective options to manage individuals as they transition to a secondary progressive phase. The current review highlights the molecular and cellular sequelae that have been identified as cooperating with and/or contributing to neurodegeneration that characterizes individuals with progressive forms of MS. We emphasize the need for appropriate monitoring via known and novel molecular and imaging biomarkers that can accurately detect and predict progression for the purposes of newly designed clinical trials that can demonstrate the efficacy of neuroprotection and potentially neurorepair. To achieve neurorepair, we focus on the modifications required in the reactive cellular and extracellular milieu in order to enable endogenous cell growth as well as transplanted cells that can integrate and/or renew the degenerative MS plaque.
Collapse
Affiliation(s)
- Olivia Ellen
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Danica Nheu
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| |
Collapse
|
13
|
Zhang X, Ma L, Liang D, Song B, Chen J, Huang Y, Xu L, Zhao P, Wu W, Zhang N, Xue R. Neurofilament Light Protein Predicts Disease Progression in Idiopathic REM Sleep Behavior Disorder. JOURNAL OF PARKINSON'S DISEASE 2023:JPD223519. [PMID: 37182898 DOI: 10.3233/jpd-223519] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Idiopathic rapid eye movement sleep behavior disorder (iRBD) is increasingly recognized as a manifestation preceding the α-synucleinopathies like Parkinson's disease (PD). Neurofilament light chain (NfL) have been reported to be higher in synucleinopathies as a sign of neurodegeneration. OBJECTIVE To evaluate whether plasma NfL is valuable in reflecting cognitive and motor status in iRBD and PD with a premorbid history of RBD (PDRBD), and predicting disease progression in iRBD. METHODS Thirty-one patients with iRBD, 30 with PDRBD, and 18 healthy controls were included in the cross-sectional and prospective study. Another cohort from the Parkinson's Progression Markers Initiative (PPMI) dataset was enrolled for verification analysis. All patients received evaluations of cognitive, motor, and autonomic function by a battery of clinical tests at baseline and follow-up. Blood NfL was measured by the Quanterix Simoa HD-1. RESULTS In our cohort, 26 patients with iRBD completed the follow-up evaluations, among whom eight (30.8%) patients displayed phenoconversion. Baseline plasma NfL cutoff value of 22.93 pg/mL performed best in distinguishing the iRBD converters from non-converters (sensitivity: 75.0%, specificity: 83.3%, area under the curve: 0.84). Cognitive and motor function were significantly correlated with NfL levels in PDRBD (correlation coefficients: -0.379, 0.399; respectively). Higher baseline NfL levels in iRBD were significantly associated with higher risks for cognitive, motor, autonomic function progression, and phenoconversion at follow-up (hazard ratios: 1.069, 1.065, 1.170, 1.065; respectively). The findings were supported by the PPMI dataset. CONCLUSION Plasma NfL is valuable in reflecting disease severity of PDRBD and predicting disease progression and phenoconversion in iRBD.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Li Ma
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Danqi Liang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingxin Song
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingshan Chen
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaqin Huang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Wu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Xue
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Sen MK, Hossain MJ, Mahns DA, Brew BJ. Validity of serum neurofilament light chain as a prognostic biomarker of disease activity in multiple sclerosis. J Neurol 2023; 270:1908-1930. [PMID: 36520240 DOI: 10.1007/s00415-022-11507-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating and neuroinflammatory disease of the human central nervous system with complex pathoetiology, heterogeneous presentations and an unpredictable course of disease progression. There remains an urgent need to identify and validate a biomarker that can reliably predict the initiation and progression of MS as well as identify patient responses to disease-modifying treatments/therapies (DMTs). Studies exploring biomarkers in MS and other neurodegenerative diseases currently focus mainly on cerebrospinal fluid (CSF) analyses, which are invasive and impractical to perform on a repeated basis. Recent studies, replacing CSF with peripheral blood samples, have revealed that the elevation of serum neurofilament light chain (sNfL) in the clinical stages of MS is, potentially, an ideal prognostic biomarker for predicting disease progression and for possibly guiding treatment decisions. However, there are unresolved factors (the definition of abnormal values of sNfL concentration, the standardisation of measurement and the amount of change in sNfL concentration that is significant) that are preventing its use as a biomarker in routine clinical practice for MS. This updated review critiques these recent findings and highlights areas for focussed work to facilitate the use of sNfL as a prognostic biomarker in MS management.
Collapse
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
- Peter Duncan Neuroscience Research Unit, St Vincent's Centre for Applied Medical Research, Darlinghurst, Sydney, 2010, Australia
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Md Jakir Hossain
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - David A Mahns
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Bruce J Brew
- Peter Duncan Neuroscience Research Unit, St Vincent's Centre for Applied Medical Research, Darlinghurst, Sydney, 2010, Australia.
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia.
- Department of Neurology, St Vincent's Hospital, Darlinghurst, 2010, Australia.
| |
Collapse
|
15
|
Abstract
This review aimed to elucidate protein biomarkers in body fluids, such as blood and cerebrospinal fluid (CSF), to identify those that may be used for early diagnosis of multiple sclerosis (MS), prediction of disease activity, and monitoring of treatment response among MS patients. The potential biomarkers elucidated in this review include neurofilament proteins (NFs), glial fibrillary acidic protein (GFAP), leptin, brain-derived neurotrophic factor (BDNF), chitinase-3-like protein 1 (CHI3L1), C-X-C motif chemokine 13 (CXCL13), and osteopontin (OPN), with each biomarker playing a different role in MS. GFAP, leptin, and CHI3L1 levels were increased in MS patient groups compared to the control group. NFs are the most studied proteins in the MS field, and significant correlations with disease activity, future progression, and treatment outcomes are evident. GFAP CSF level shows a different pattern by MS subtype. Increased concentration of CHI3L1 in the blood/CSF of clinically isolated syndrome (CIS) is an independent predictive factor of conversion to definite MS. BDNF may be affected by chronic progression of MS. CHI3L1 has potential as a biomarker for early diagnosis of MS and prediction of disability progression, while CXCL13 has potential as a biomarker of prognosis of CIS and reflects MS disease activity. OPN was an indicator of disease severity. A periodic detailed patient evaluation should be performed for MS patients, and broadly and easily accessible biomarkers with higher sensitivity and specificity in clinical settings should be identified.
Collapse
Affiliation(s)
- Jun-Soon Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Buchmann A, Pirpamer L, Pinter D, Voortman M, Helmlinger B, Pichler A, Maceski AM, Benkert P, Bachmaier G, Ropele S, Reindl M, Leppert D, Kuhle J, Enzinger C, Khalil M. High serum neurofilament light chain levels correlate with brain atrophy and physical disability in multiple sclerosis. Eur J Neurol 2023; 30:1389-1399. [PMID: 36779855 DOI: 10.1111/ene.15742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/23/2022] [Accepted: 02/02/2023] [Indexed: 02/14/2023]
Abstract
BACKGROUND AND PURPOSE Serum neurofilament light chain (sNfL) is a promising biomarker of neuroaxonal damage in persons with multiple sclerosis (pwMS). In cross-sectional studies, sNfL has been associated with disease activity and brain magnetic resonance imaging (MRI) changes; however, it is still unclear to what extent in particular high sNfL levels impact on subsequent disease evolution. METHODS sNfL was quantified by an ultrasensitive single molecule array (Simoa) in 199 pwMS (median age = 34.2 years, 64.3% female) and 49 controls. All pwMS underwent 3-T MRI to assess global and compartmental normalized brain volumes, T2-lesion load, and cortical mean thickness. Follow-up data and serum samples were available in 144 pwMS (median follow-up time = 3.8 years). Linear and binary logistic models were used to estimate the independent contribution of sNfL for changes in MRI and Expanded Disability Status Scale (EDSS). Age-corrected sNfL z-scores from a normative database of healthy controls were used for sensitivity analyses. RESULTS High sNfL levels at baseline were associated with atrophy measures of the whole brain (standardized beta coefficient βj = -0.352, p < 0.001), white matter (βj = -0.229, p = 0.007), thalamus (βj = -0.372, p = 0.004), and putamen (βj = -1.687, p = 0.012). pwMS with high levels of sNfL at baseline and follow-up had a greater risk of EDSS worsening (p = 0.007). CONCLUSIONS Already single time point elevation of sNfL has a distinct effect on brain volume changes over a short-term period, and repeated high levels of sNfL indicate accumulating physical disability. Serial assessment of sNfL may provide added value in the clinical management of pwMS.
Collapse
Affiliation(s)
| | - Lukas Pirpamer
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Daniela Pinter
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | | | | | - Aleksandra Maleska Maceski
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.,Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.,Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Gerhard Bachmaier
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - David Leppert
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.,Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.,Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Christian Enzinger
- Department of Neurology, Medical University of Graz, Graz, Austria.,Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, Graz, Austria
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| |
Collapse
|
17
|
Højsgaard Chow H, Talbot J, Lundell H, Marstrand L, Gøbel Madsen C, Bach Søndergaard H, Bredahl Hansen M, Solberg Sørensen P, Siebner HR, Sellebjerg F. Dimethyl fumarate treatment of primary progressive multiple sclerosis: results of an open-label extension study. Mult Scler Relat Disord 2023; 70:104458. [PMID: 36586351 DOI: 10.1016/j.msard.2022.104458] [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: 11/16/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Dimethyl fumarate treatment is approved in Europe for patients with relapsing-remitting multiple sclerosis (MS) and in the US for relapsing forms of MS. We recently published the results of the first randomized placebo-controlled trial of 48 weeks of treatment with dimethyl fumarate or placebo in primary progressive MS (PPMS) (clinicaltrial.gov NCT02959658). The placebo-controlled phase of the trial did not meet its primary endpoint (reduction in cerebrospinal fluid concentrations of neurofilament light chain [NFL]). AIM To investigate the effects of dimethyl fumarate treatment in the open-label extension phase of the trial (week 48-96), where all patients were treated with DMF. METHODS Reported data are from screening, week 48, and week 96 visits. Patients were clinically evaluated with Expanded Disability Status Scale (EDSS), 9-Hole Peg Test (9HPT), Timed 25-Foot Walk (T25FW) test, Symbol Digit Modalities Test (SDMT), California Verbal Learning Test, and Brief Visuospatial Memory-Revised. Serum NFL concentrations were measured by single-molecule array analysis. MRI was performed on a 3 tesla MRI scanner and included: new/enlarging lesions, volume of lesions, cortical grey matter, putamen, thalamus, and normal-appearing white matter, and additional diffusion tensor imaging and magnetization transfer ratio measures. RESULTS Forty-two patients entered the open-label treatment phase, and 33 patients (61%) had complete data sets at week 96. The remaining 39% did not complete the trial and were not evaluated at week 96. We found no evidence of differences in clinical and MRI measures between patients initially treated with dimethyl fumarate and patients initially treated with placebo from baseline to week 48 and from week 48-96, where all patients were treated with dimethyl fumarate. Serum NFL concentrations remained stable in both groups over 96 weeks. Assessed with either EDSS, T25FW, or 9HPT at week 96, progression was observed for 14 patients (45%). Interestingly, another 15 patients (46%) had improvement in one or more of these domains. Applying a cut-off of 8 points, 2 (6%) patients worsened on SDMT, 25 (78%) did not change, and 5 (16%) improved. CONCLUSIONS Dimethyl fumarate treatment showed no effects on neither clinical nor MRI outcomes or changes in serum concentrations of NFL. An expected number of patients showed evidence of progression on standard clinical scales; however, this was matched by an equal number of patients improving. The reasons for the physical improvement in an unexpectedly high proportion of patients must be addressed in future studies.
Collapse
Affiliation(s)
- Helene Højsgaard Chow
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark.
| | - Jacob Talbot
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark
| | - Henrik Lundell
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen Denmark, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Lisbet Marstrand
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark
| | - Camilla Gøbel Madsen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen Denmark, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Helle Bach Søndergaard
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark
| | - Malene Bredahl Hansen
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark
| | - Per Solberg Sørensen
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen Denmark, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Nordre Ringvej 57, 2600 Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Copenhagen, Denmark
| |
Collapse
|
18
|
Neurofilament light chains in serum as biomarkers of axonal damage in early MS lesions: a histological-serological correlative study. J Neurol 2023; 270:1416-1429. [PMID: 36372867 PMCID: PMC9971126 DOI: 10.1007/s00415-022-11468-2] [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: 09/19/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease associated with axonal injury, and neurofilament light chains in serum (sNfL) are considered a biomarker for this damage. We aimed to investigate the relationship between sNfL and the axonal damage in early MS lesions in a special cohort of biopsied patients. sNfL from 106 biopsied patients with 26 follow-up samples were analyzed using single-molecule array (SiMoA) technology. Findings were correlated with clinical parameters and histological findings of acute axonal damage (APP-positive spheroids) and axonal loss in different lesion stages. A median of 59 pg/ml sNfL was found (range 8-3101 pg/ml). sNfL levels correlated with APP-positive spheroids in early active demyelinating lesions that represent the earliest lesion stages (p < 0.01). A significant negative correlation between sNfL levels in follow-up blood samples and axonal density in normal-appearing white matter was also observed (p = 0.02). sNfL levels correlated with the Expanded Disability Status Score at biopsy (p < 0.01, r = 0.49) and at last clinical follow-up (p < 0.01, r = 0.66). In conclusion, sNfL likely represent a compound measure of recent and ongoing neuroaxonal damage. We found that sNfL in biopsied MS patients correlate with acute axonal damage in the earliest MS lesion stages. Determination of sNfL levels thus allows insight into brain pathology and underlines the relevance of relapse-associated lesional pathology. Axonal loss in normal-appearing white matter contributes to sNfL levels independent of relapses. Since sNfL levels correlate with clinical disability, they may predict the future disability of patients and help with individual treatment decisions.
Collapse
|
19
|
Ning L, Wang B. Neurofilament light chain in blood as a diagnostic and predictive biomarker for multiple sclerosis: A systematic review and meta-analysis. PLoS One 2022; 17:e0274565. [PMID: 36103562 PMCID: PMC9473405 DOI: 10.1371/journal.pone.0274565] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Neurofilament light chain (NfL) in cerebrospinal fluid (CSF) is a biomarker of multiple sclerosis (MS). However, CSF sampling is invasive and has limited the clinical application. With the development of highly sensitive single-molecule assay, the accurate quantification of the very low NfL levels in blood become feasible. As evidence being accumulated, we performed a meta-analysis to evaluate the diagnostic and predictive value of blood NfL in MS patients.
Methods
We performed literature search on PubMed, EMBASE, Web of Science and Cochrane Library from inception to May 31, 2022. The blood NfL differences between MS vs. controls, MS vs. clinically isolated syndrome (CIS), progressive MS (PMS) vs. relapsing-remitting MS (RRMS), and MS in relapse vs. MS in remission were estimated by standard mean difference (SMD) and corresponding 95% confidence interval (CI). Pooled hazard ratio (HR) and 95%CI were calculated to predict time to reach Expanded Disability Status Scale (EDSS) score≥4.0 and to relapse.
Results
A total of 28 studies comprising 6545 MS patients and 2477 controls were eligible for meta-analysis of diagnosis value, and 5 studies with 4444 patients were synthesized in analysis of predictive value. Blood NfL levels were significantly higher in MS patients vs. age-matched controls (SMD = 0.64, 95%CI 0.44–0.85, P<0.001), vs. non-matched controls (SMD = 0.76, 95%CI 0.56–0.96, P<0.001) and vs. CIS patients (SMD = 0.30, 95%CI 0.18–0.42, P<0.001), in PMS vs. RRMS (SMD = 0.56, 95%CI 0.27–0.85, P<0.001), and in relapsed patients vs. remitted patients (SMD = 0.54, 95%CI 0.16–0.92, P = 0.005). Patients with high blood NfL levels had shorter time to reach EDSS score≥4.0 (HR = 2.36, 95%CI 1.32–4.21, P = 0.004) but similar time to relapse (HR = 1.32, 95%CI 0.90–1.93, P = 0.155) compared to those with low NfL levels.
Conclusion
As far as we know, this is the first meta-analysis evaluating the diagnosis and predictive value of blood NfL in MS. The present study indicates blood NfL may be a useful biomarker in diagnosing MS, distinguishing MS subtypes and predicting disease worsening in the future.
Collapse
Affiliation(s)
- Liangxia Ning
- Department of Neurology, Yuncheng Central Hospital, The Eighth Shanxi Medical University, Yuncheng, China
| | - Bin Wang
- Department of Neurology, Yuncheng Central Hospital, The Eighth Shanxi Medical University, Yuncheng, China
- * E-mail:
| |
Collapse
|
20
|
Rise HH, Brune S, Chien C, Berge T, Bos SD, Andorrà M, Valdeolivas IP, Beyer MK, Sowa P, Scheel M, Brandt AU, Asseyer S, Blennow K, Pedersen ML, Zetterberg H, de Schotten MT, Cellerino M, Uccelli A, Paul F, Villoslada P, Harbo HF, Westlye LT, Høgestøl EA. Brain disconnectome mapping derived from white matter lesions and serum neurofilament light levels in multiple sclerosis: A longitudinal multicenter study. Neuroimage Clin 2022; 35:103099. [PMID: 35772194 PMCID: PMC9253471 DOI: 10.1016/j.nicl.2022.103099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Connectivity-based approaches incorporating the distribution and magnitude of the extended brain network aberrations caused by lesions may offer higher sensitivity for axonal damage in patients with multiple sclerosis (MS) than conventional lesion characteristics. Using individual brain disconnectome mapping, we tested the longitudinal associations between putative imaging-based brain network aberrations and levels of serum neurofilament light chain (NfL) as a neuroaxonal injury biomarker. METHODS MS patients (n = 312, mean age 42.9 years, 71 % female) and healthy controls (HC) (n = 59, mean age 39.9 years, 78 % female) were prospectively enrolled at four European MS centres, and reassessed after two years (MS, n = 242; HC, n = 30). Post-processing of 3 Tesla (3 T) MRI data was performed at one centre using a harmonized pipeline, and disconnectome maps were calculated using BCBtoolkit based on individual lesion maps. Global disconnectivity (GD) was defined as the average disconnectome probability in each patient's white matter. Serum NfL concentrations were measured by single molecule array (Simoa). Robust linear mixed models (rLMM) with GD or T2-lesion volume (T2LV) as dependent variables, patient as a random factor, serum NfL, age, sex, timepoint for visit, diagnosis, treatment, and center as fixed factors were run. RESULTS rLMM revealed significant associations between GD and serum NfL (t = 2.94, p = 0.003), age (t = 4.21, p = 2.5 × 10-5), and longitudinal changes in NfL (t = -2.29, p = 0.02), but not for sex (t = 0.63, p = 0.53) or treatments (t = 0.80-0.83, p = 0.41-0.42). Voxel-wise analyses revealed significant associations between dysconnectivity in cerebellar and brainstem regions and serum NfL (t = 7.03, p < 0.001). DISCUSSION In our prospective multi-site MS cohort, rLMMs demonstrated that the extent of global and regional brain disconnectivity is sensitive to a systemic biomarker of axonal damage, serum NfL, in patients with MS. These findings provide a neuroaxonal correlate of advanced disconnectome mapping and provide a platform for further investigations of the functional and potential clinical relevance of brain disconnectome mapping in patients with brain disorders.
Collapse
Affiliation(s)
- Henning H Rise
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Synne Brune
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Claudia Chien
- Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin & Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Experimental and Clinical Research Center, Germany; Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, Germany; Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department for Psychiatry and Psychotherapy, Germany
| | - Tone Berge
- Department of Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway; Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway
| | - Steffan D Bos
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Magí Andorrà
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | | | - Mona K Beyer
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Piotr Sowa
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Michael Scheel
- Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, Germany; Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Neuroradiology, Germany
| | - Alexander U Brandt
- Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin & Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Experimental and Clinical Research Center, Germany; Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, Germany; Department of Neurology, University of California, Irvine, CA, USA
| | - Susanna Asseyer
- Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin & Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Experimental and Clinical Research Center, Germany
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Mads L Pedersen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France; Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives- UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France
| | - Maria Cellerino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Friedemann Paul
- Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin & Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Experimental and Clinical Research Center, Germany; Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, Germany; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pablo Villoslada
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Hanne F Harbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway; KG Jebsen, Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Einar A Høgestøl
- Department of Psychology, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
21
|
Razia R, Majeed F, Amin R, Mukhtar S, Mehmood K, Baig DN. The analysis of dynamic gene expression patterns in peripheral blood of multiple sclerosis patients indicates possible diagnostic and prognostic biomarkers. Mol Immunol 2022; 147:147-156. [PMID: 35594733 DOI: 10.1016/j.molimm.2022.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/08/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Among numerous invasive procedures for the research of biomarkers, blood-based indicators are regarded as marginally non-invasive procedures in the diagnosis and prognosis of demyelinating disorders, including multiple sclerosis (MS). In this study, we looked into the blood-derived gene expression profiles of patients with multiple sclerosis to investigate their clinical traits and linked them with dysregulated gene expressions to establish diagnostic and prognostic indicators. METHODS We included 51 patients with relapsing-remitting MS (RRMS, n = 31), clinically isolated syndrome (CIS, n = 12), primary progressive MS (PPMS, n = 8) and a control group (n = 51). Using correlational analysis, the transcriptional patterns of chosen gene panels were examined and subsequently related with disease duration and the expanded disease disability score (EDSS). In addition, principal component analysis, univariate regression, and logistic regression analysis were employed to highlight distinct profiles of genes and prognosticate the excellent biomarkers of this illness. RESULTS Our findings demonstrated that neurofilament light (NEFL), tumor necrosis factor α (TNF-α), Tau, and clusterin (CLU) were revealed to be increased in recruited patients, whereas the presenilin-1 (PSEN1) and cell-surface glycoprotein-44 (CD44) were downregulated. Principal Component Analysis revealed distinct patterns between the MS and control groups. Correlation analysis indicated co-dependent dysregulated genes and their differential expression with clinical findings. Furthermore, logistic regression demonstrated that Clusterin (AUC=0.940), NEFL (AUC=0.775), TNF-α (AUC=0.817), Tau (AUC=0.749), PSEN1 (AUC=0.6913), and CD44 (AUC=0.832) had diagnostic relevance. Following the univariate linear regression, a significant regression equation was found between EDSS and IGF-1 (R2 adj = 0.10844; p= 0.0060), APP (R2 adj = 0.1107; p= 0.0098), and PSEN1 (R2 adj = 0.1266; p=0.0102). CONCLUSION This study exhibits dynamic gene expression patterns that represent the significance of specified genes that are prospective diagnostic and prognostic biomarkers for multiple sclerosis.
Collapse
Affiliation(s)
- Rabat Razia
- School of Life Sciences, Forman Christian College (A Chartered University), Lahore 54600, Pakistan.
| | | | - Rehab Amin
- School of Life Sciences, Forman Christian College (A Chartered University), Lahore 54600, Pakistan.
| | - Shahid Mukhtar
- Punjab Institute of Neurosciences, Ferozpur Road, Lahore 54000, Punjab
| | - Khalid Mehmood
- Punjab Institute of Neurosciences, Ferozpur Road, Lahore 54000, Punjab
| | - Deeba Noreen Baig
- School of Life Sciences, Forman Christian College (A Chartered University), Lahore 54600, Pakistan.
| |
Collapse
|
22
|
Biernacki T, Kokas Z, Sandi D, Füvesi J, Fricska-Nagy Z, Faragó P, Kincses TZ, Klivényi P, Bencsik K, Vécsei L. Emerging Biomarkers of Multiple Sclerosis in the Blood and the CSF: A Focus on Neurofilaments and Therapeutic Considerations. Int J Mol Sci 2022; 23:ijms23063383. [PMID: 35328802 PMCID: PMC8951485 DOI: 10.3390/ijms23063383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/12/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Multiple Sclerosis (MS) is the most common immune-mediated chronic neurodegenerative disease of the central nervous system (CNS) affecting young people. This is due to the permanent disability, cognitive impairment, and the enormous detrimental impact MS can exert on a patient's health-related quality of life. It is of great importance to recognise it in time and commence adequate treatment at an early stage. The currently used disease-modifying therapies (DMT) aim to reduce disease activity and thus halt disability development, which in current clinical practice are monitored by clinical and imaging parameters but not by biomarkers found in blood and/or the cerebrospinal fluid (CSF). Both clinical and radiological measures routinely used to monitor disease activity lack information on the fundamental pathophysiological features and mechanisms of MS. Furthermore, they lag behind the disease process itself. By the time a clinical relapse becomes evident or a new lesion appears on the MRI scan, potentially irreversible damage has already occurred in the CNS. In recent years, several biomarkers that previously have been linked to other neurological and immunological diseases have received increased attention in MS. Additionally, other novel, potential biomarkers with prognostic and diagnostic properties have been detected in the CSF and blood of MS patients. AREAS COVERED In this review, we summarise the most up-to-date knowledge and research conducted on the already known and most promising new biomarker candidates found in the CSF and blood of MS patients. DISCUSSION the current diagnostic criteria of MS relies on three pillars: MRI imaging, clinical events, and the presence of oligoclonal bands in the CSF (which was reinstated into the diagnostic criteria by the most recent revision). Even though the most recent McDonald criteria made the diagnosis of MS faster than the prior iteration, it is still not an infallible diagnostic toolset, especially at the very early stage of the clinically isolated syndrome. Together with the gold standard MRI and clinical measures, ancillary blood and CSF biomarkers may not just improve diagnostic accuracy and speed but very well may become agents to monitor therapeutic efficacy and make even more personalised treatment in MS a reality in the near future. The major disadvantage of these biomarkers in the past has been the need to obtain CSF to measure them. However, the recent advances in extremely sensitive immunoassays made their measurement possible from peripheral blood even when present only in minuscule concentrations. This should mark the beginning of a new biomarker research and utilisation era in MS.
Collapse
Affiliation(s)
- Tamás Biernacki
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Zsófia Kokas
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Dániel Sandi
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Judit Füvesi
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Zsanett Fricska-Nagy
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Péter Faragó
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Tamás Zsigmond Kincses
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
- Albert Szent-Györgyi Clinical Centre, Department of Radiology, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
| | - Péter Klivényi
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - Krisztina Bencsik
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
| | - László Vécsei
- Albert Szent-Györgyi Clinical Centre, Department of Neurology, Faculty of General Medicine, University of Szeged, 6725 Szeged, Hungary; (T.B.); (Z.K.); (D.S.); (J.F.); (Z.F.-N.); (P.F.); (T.Z.K.); (P.K.); (K.B.)
- MTA-SZTE Neuroscience Research Group, University of Szeged, 6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-356; Fax: +36-62-545-597
| |
Collapse
|
23
|
Wendel EM, Bertolini A, Kousoulos L, Rauchenzauner M, Schanda K, Wegener-Panzer A, Baumann M, Reindl M, Otto M, Rostásy K. Serum neurofilament light-chain levels in children with monophasic myelin oligodendrocyte glycoprotein-associated disease, multiple sclerosis, and other acquired demyelinating syndrome. Mult Scler 2022; 28:1553-1561. [DOI: 10.1177/13524585221081090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: To assess the diagnostic and prognostic potential of serum neurofilament light chain (sNfL) in children with first acquired demyelinating syndrome (ADS). Methods: We selected 129 children with first ADS including 19 children with myelin oligodendrocyte glycoprotein (MOG)-antibody associated disease (MOGAD), 36 MOG/AQP4-seronegative ADS, and 74 with multiple sclerosis (MS) from the BIOMARKER study cohort. All children had a complete set of clinical, radiological, laboratory data and serum for NfL measurement using a highly sensitive digital ELISA (SIMOA). A control group of 35 children with non-inflammatory neurological diseases was included. sNfL levels were compared across patient groups according to clinical, laboratory, neuroradiological features and outcome after 2 years. Results: sNfL levels were significantly increased in MOGAD, seronegative ADS and MS compared to controls ( p-value < 0.001), in particular in children with an acute disseminated encephalomyelitis (ADEM)-like magnetic resonance imaging (MRI) pattern ( p < 0.001) or longitudinally extensive myelitis ( p < 0.01). In pediatric MS, elevated sNfL levels were significantly associated with higher numbers of cerebral ( p < 0.001) and presence of spinal ( p < 0.05) MRI lesions at baseline and predicted a higher number of relapses ( p < 0.05). Conclusion: sNfL levels are significantly elevated in all three studied pediatric ADS subtypes indicating neuroaxonal injury. In pediatric MS high levels of sNfL are associated with risk factors for disease progression.
Collapse
Affiliation(s)
- Eva-Maria Wendel
- Department of Pediatrics, Olgahospital, Klinikum Stuttgart, Stuttgart, Germany
| | - Annikki Bertolini
- Department of Pediatric Neurology, Children’s Hospital Datteln, University Witten/Herdecke, Datteln, Germany
| | - Lampros Kousoulos
- Department of Pediatric Neurology, Children’s Hospital Datteln, University Witten/Herdecke, Datteln, Germany
| | - Markus Rauchenzauner
- Department of Pediatrics and Neonatology, Kliniken Ostallgäu-Kaufbeuren, Kaufbeuren, Germany/Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Kathrin Schanda
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Wegener-Panzer
- Department of Pediatric Radiology, Children`s Hospital Datteln, University Witten/Herdecke, Datteln, Germany
| | - Matthias Baumann
- Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Otto
- Department of Neurology, University Hospital of Ulm, Ulm, Germany; Department of Neurology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Kevin Rostásy
- Department of Pediatric Neurology, Children’s Hospital Datteln, University Witten/Herdecke, Datteln, Germany
| |
Collapse
|
24
|
Welton T, Tan YJ, Saffari SE, Ng SYE, Chia NSY, Yong ACW, Choi X, Heng DL, Shih YC, Hartono S, Lee W, Xu Z, Tay KY, Au WL, Tan EK, Chan LL, Ng ASL, Tan LCS. Plasma Neurofilament Light Concentration Is Associated with Diffusion-Tensor MRI-Based Measures of Neurodegeneration in Early Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:2135-2146. [PMID: 36057833 DOI: 10.3233/jpd-223414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Neurofilament light is a marker of axonal degeneration, whose measurement from peripheral blood was recently made possible by new assays. OBJECTIVE We aimed to determine whether plasma neurofilament light chain (NfL) concentration reflects brain white matter integrity in patients with early Parkinson's disease (PD). METHODS 137 early PD patients and 51 healthy controls were included. Plasma NfL levels were measured using ultrasensitive single molecule array. 3T MRI including diffusion tensor imaging was acquired for voxelwise analysis of association between NfL and both fractional anisotropy (FA) and mean diffusivity (MD) in white matter tracts and subcortical nuclei. RESULTS A pattern of brain microstructural changes consistent with neurodegeneration was associated with increased plasma NfL in most of the frontal lobe and right internal capsule, with decreased FA and increased MD. The same clusters were also associated with poorer global cognition. A significant cluster in the left putamen was associated with increased NfL, with a significantly greater effect in PD than controls. CONCLUSION Plasma NfL may be associated with brain microstructure, as measured using diffusion tensor imaging, in patients with early PD. Higher plasma NfL was associated with a frontal pattern of neurodegeneration that also correlates with cognitive performance in our cohort. This may support a future role for plasma NfL as an accessible biomarker for neurodegeneration and cognitive dysfunction in PD.
Collapse
Affiliation(s)
- Thomas Welton
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Neuroscience Academic Clinical Program, Duke-NUS Medical School, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Seyed Ehsan Saffari
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Samuel Y E Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Nicole S Y Chia
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Alisa C W Yong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Xinyi Choi
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
| | - Dede Liana Heng
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
| | - Yao-Chia Shih
- Radiological Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan City, Taiwan
| | - Septian Hartono
- Neuroscience Academic Clinical Program, Duke-NUS Medical School, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
| | - Weiling Lee
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore
| | - Zheyu Xu
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Kay Yaw Tay
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Wing Lok Au
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
| | - Ling Ling Chan
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Radiological Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore
- Neuroscience and Behavioural Disorders Program, Duke-NUS Medical School, Singapore
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
- Neuroscience and Behavioural Disorders Program, Duke-NUS Medical School, Singapore
| | - Louis C S Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
- Parkinson Disease and Movement Disorders Centre, Parkinson Foundation Center of Excellence, National Neuroscience Institute, Singapore, Tan Tock Seng Hospital, Singapore
| |
Collapse
|
25
|
Neuroprotective Effect of Glatiramer Acetate on Neurofilament Light Chain Leakage and Glutamate Excess in an Animal Model of Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms222413419. [PMID: 34948217 PMCID: PMC8707261 DOI: 10.3390/ijms222413419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Axonal and neuronal pathologies are a central constituent of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), induced by the myelin oligodendrocyte glycoprotein (MOG) 35–55 peptide. In this study, we investigated neurodegenerative manifestations in chronic MOG 35–55 induced EAE and the effect of glatiramer acetate (GA) treatment on these manifestations. We report that the neuronal loss seen in this model is not attributed to apoptotic neuronal cell death. In EAE-affected mice, axonal damage prevails from the early disease phase, as revealed by analysis of neurofilament light (NFL) leakage into the sera along the disease duration, as well as by immunohistological examination. Elevation of interstitial glutamate concentrations measured in the cerebrospinal fluid (CSF) implies that glutamate excess plays a role in the damage processes inflicted by this disease. GA applied as a therapeutic regimen to mice with apparent clinical symptoms significantly reduces the pathological manifestations, namely apoptotic cell death, NFL leakage, histological tissue damage, and glutamate excess, thus corroborating the neuroprotective consequences of this treatment.
Collapse
|
26
|
Yuan A, Nixon RA. Neurofilament Proteins as Biomarkers to Monitor Neurological Diseases and the Efficacy of Therapies. Front Neurosci 2021; 15:689938. [PMID: 34646114 PMCID: PMC8503617 DOI: 10.3389/fnins.2021.689938] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Biomarkers of neurodegeneration and neuronal injury have the potential to improve diagnostic accuracy, disease monitoring, prognosis, and measure treatment efficacy. Neurofilament proteins (NfPs) are well suited as biomarkers in these contexts because they are major neuron-specific components that maintain structural integrity and are sensitive to neurodegeneration and neuronal injury across a wide range of neurologic diseases. Low levels of NfPs are constantly released from neurons into the extracellular space and ultimately reach the cerebrospinal fluid (CSF) and blood under physiological conditions throughout normal brain development, maturation, and aging. NfP levels in CSF and blood rise above normal in response to neuronal injury and neurodegeneration independently of cause. NfPs in CSF measured by lumbar puncture are about 40-fold more concentrated than in blood in healthy individuals. New ultra-sensitive methods now allow minimally invasive measurement of these low levels of NfPs in serum or plasma to track disease onset and progression in neurological disorders or nervous system injury and assess responses to therapeutic interventions. Any of the five Nf subunits - neurofilament light chain (NfL), neurofilament medium chain (NfM), neurofilament heavy chain (NfH), alpha-internexin (INA) and peripherin (PRPH) may be altered in a given neuropathological condition. In familial and sporadic Alzheimer's disease (AD), plasma NfL levels may rise as early as 22 years before clinical onset in familial AD and 10 years before sporadic AD. The major determinants of elevated levels of NfPs and degradation fragments in CSF and blood are the magnitude of damaged or degenerating axons of fiber tracks, the affected axon caliber sizes and the rate of release of NfP and fragments at different stages of a given neurological disease or condition directly or indirectly affecting central nervous system (CNS) and/or peripheral nervous system (PNS). NfPs are rapidly emerging as transformative blood biomarkers in neurology providing novel insights into a wide range of neurological diseases and advancing clinical trials. Here we summarize the current understanding of intracellular NfP physiology, pathophysiology and extracellular kinetics of NfPs in biofluids and review the value and limitations of NfPs and degradation fragments as biomarkers of neurodegeneration and neuronal injury.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
- Department of Cell Biology, New York University Grossman School of Medicine, (NYU), Neuroscience Institute, New York, NY, United States
| |
Collapse
|
27
|
Lin TY, Vitkova V, Asseyer S, Martorell Serra I, Motamedi S, Chien C, Ditzhaus M, Papadopoulou A, Benkert P, Kuhle J, Bellmann-Strobl J, Ruprecht K, Paul F, Brandt AU, Zimmermann HG. Increased Serum Neurofilament Light and Thin Ganglion Cell-Inner Plexiform Layer Are Additive Risk Factors for Disease Activity in Early Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/5/e1051. [PMID: 34348969 PMCID: PMC8362351 DOI: 10.1212/nxi.0000000000001051] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/10/2021] [Indexed: 11/29/2022]
Abstract
Objective To investigate the association of combined serum neurofilament light chain (sNfL) and retinal optical coherence tomography (OCT) measurements with future disease activity in patients with early multiple sclerosis (MS). Methods We analyzed sNfL by single molecule array technology and performed OCT measurements in a prospective cohort of 78 patients with clinically isolated syndrome and early relapsing-remitting MS with a median (interquartile range) follow-up of 23.9 (23.3–24.7) months. Patients were grouped into those with abnormal or normal sNfL levels, defined as sNfL ≥/<80th percentile of age-corrected reference values. Likewise, patients were grouped by a median split into those with thin or thick ganglion cell and inner plexiform layer (GCIP), peripapillary retinal nerve fiber layer, and inner nuclear layer in nonoptic neuritis eyes. Outcome parameters were violation of no evidence of disease activity (NEDA-3) criteria or its components. Results Patients with abnormal baseline sNfL had a higher risk of violating NEDA-3 (hazard ratio [HR] 2.28, 95% CI 1.27–4.09, p = 0.006) and developing a new brain lesion (HR 2.47, 95% CI 1.30–4.69, p = 0.006), but not for a new relapse (HR 2.21, 95% CI 0.97–5.03, p = 0.058). Patients with both abnormal sNfL and thin GCIP had an even higher risk for NEDA-3 violation (HR 3.61, 95% CI 1.77–7.36, p = 4.2e−4), new brain lesion (HR 3.19, 95% CI 1.51–6.76, p = 0.002), and new relapse (HR 5.38, 95% CI 1.61–17.98, p = 0.006) than patients with abnormal sNfL alone. Conclusions In patients with early MS, the presence of both abnormal sNfL and thin GCIP is a stronger risk factor for future disease activity than the presence of each parameter alone.
Collapse
Affiliation(s)
- Ting-Yi Lin
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Viktoriya Vitkova
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Susanna Asseyer
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Ivette Martorell Serra
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Seyedamirhosein Motamedi
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Claudia Chien
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Marc Ditzhaus
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Athina Papadopoulou
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Pascal Benkert
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Jens Kuhle
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Judith Bellmann-Strobl
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Klemens Ruprecht
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Friedemann Paul
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Alexander U Brandt
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine
| | - Hanna G Zimmermann
- From the Experimental and Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Max-Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; NeuroCure Clinical Research Center (T.-Y.L., V.V., S.A., I.M.S., S.M., C.C., A.P., J.B.-S., F.P., A.U.B., H.G.Z.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Psychiatry and Psychotherapy (C.C.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Department of Statistics (M.D.), TU Dortmund University, Germany; Neurology Clinic and Policlinic (A.P., J.K.), MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel; Clinical Trial Unit (P.B.), Department of Clinical Research, University Hospital Basel, University of Basel, Switzerland; Department of Neurology (K.R., F.P.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and Department of Neurology (A.U.B.), University of California, Irvine.
| |
Collapse
|
28
|
Yun T, Koo Y, Chae Y, Lee D, Kim H, Kim S, Chang D, Na K, Yang M, Kang B. Neurofilament light chain as a biomarker of meningoencephalitis of unknown etiology in dogs. J Vet Intern Med 2021; 35:1865-1872. [PMID: 34114244 PMCID: PMC8295659 DOI: 10.1111/jvim.16184] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neurofilament light chain (NfL) is a neuron-specific cytoskeletal protein expressed in axons. Damaged axons of the central nervous system release NfLs into the cerebrospinal fluid (CSF) and the blood. In humans with neurologic diseases, NfL is used as a biomarker. OBJECTIVES To identify the potential of NfL as a supportive tool for the diagnosis, prognosis, and monitoring of meningoencephalitis of unknown etiology (MUE) in dogs. ANIMALS Twenty-six client-owned healthy dogs, 10 normal Beagle dogs, and 38 client-owned MUE dogs. METHODS Cohort study. The concentrations of NfL in serum and CSF were measured using single-molecule array technology. RESULTS Median NfL concentration was significantly higher in MUE dogs (serum, 125 pg/mL; CSF, 14 700 pg/mL) than in healthy dogs (serum, 11.8 pg/mL, P < .0001; CSF, 1410 pg/mL, P = .0002). The areas under the receiver operating characteristic curves of serum and CSF NfL concentrations were 0.99 and 0.95, respectively. The cut-off values were 41.5 pg/mL (serum) and 4005 pg/mL (CSF) for differentiating between healthy and MUE dogs, with sensitivities of 89.19% and 90%, respectively, and specificities of 96.97% and 100%, respectively. The NfL concentration showed a significant decrease (pretreatment, 122 pg/mL; posttreatment, 36.6 pg/mL; P = .02) in the good treatment-response group and a significant increase (pretreatment, 292.5 pg/mL; posttreatment, 1880 pg/mL, P = .03) in the poor treatment-response group. CONCLUSIONS AND CLINICAL IMPORTANCE Neurofilament light chain is a potential biomarker for diagnosing MUE and evaluating response to treatment.
Collapse
Affiliation(s)
- Taesik Yun
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Yoonhoi Koo
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Yeon Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Dohee Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Hakhyun Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Soochong Kim
- Laboratory of Veterinary Pathology, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Dongwoo Chang
- Section of Veterinary Medical Imaging, Veterinary Teaching Hospital, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Ki‐Jeong Na
- Laboratory of Veterinary Laboratory Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Mhan‐Pyo Yang
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| | - Byeong‐Teck Kang
- Laboratory of Veterinary Internal Medicine, College of Veterinary MedicineChungbuk National UniversityCheongjuChungbukRepublic of Korea
| |
Collapse
|
29
|
Bittner S, Oh J, Havrdová EK, Tintoré M, Zipp F. The potential of serum neurofilament as biomarker for multiple sclerosis. Brain 2021; 144:2954-2963. [PMID: 34180982 PMCID: PMC8634125 DOI: 10.1093/brain/awab241] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/27/2021] [Accepted: 06/14/2021] [Indexed: 12/03/2022] Open
Abstract
Multiple sclerosis is a highly heterogeneous disease, and the detection of neuroaxonal damage as well as its quantification is a critical step for patients. Blood-based serum neurofilament light chain (sNfL) is currently under close investigation as an easily accessible biomarker of prognosis and treatment response in patients with multiple sclerosis. There is abundant evidence that sNfL levels reflect ongoing inflammatory-driven neuroaxonal damage (e.g. relapses or MRI disease activity) and that sNfL levels predict disease activity over the next few years. In contrast, the association of sNfL with long-term clinical outcomes or its ability to reflect slow, diffuse neurodegenerative damage in multiple sclerosis is less clear. However, early results from real-world cohorts and clinical trials using sNfL as a marker of treatment response in multiple sclerosis are encouraging. Importantly, clinical algorithms should now be developed that incorporate the routine use of sNfL to guide individualized clinical decision-making in people with multiple sclerosis, together with additional fluid biomarkers and clinical and MRI measures. Here, we propose specific clinical scenarios where implementing sNfL measures may be of utility, including, among others: initial diagnosis, first treatment choice, surveillance of subclinical disease activity and guidance of therapy selection.
Collapse
Affiliation(s)
- 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
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Eva Kubala Havrdová
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Mar Tintoré
- Department of Neurology, Hospital General Universitari Vall D'Hebron, Cemcat, Barcelona, Spain
| | - Frauke Zipp
- 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
| |
Collapse
|
30
|
Saraste M, Bezukladova S, Matilainen M, Sucksdorff M, Kuhle J, Leppert D, Airas L. Increased serum glial fibrillary acidic protein associates with microstructural white matter damage in multiple sclerosis: GFAP and DTI. Mult Scler Relat Disord 2021; 50:102810. [PMID: 33556656 DOI: 10.1016/j.msard.2021.102810] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Astrocytes and microglial cells are now recognized as active players in contributing to the diffuse neuroaxonal damage associated with disease progression of multiple sclerosis (MS). The serum level of glial fibrillary acidic protein (GFAP), a biomarker for astrocytic activation, is increased in MS and associates with disease progression and disability. Similarly, diffusion tensor imaging (DTI) parameters for microstructural changes in brain, including demyelination and axonal loss, associate with disability. The association between brain DTI parameters and serum GFAP has not been previously explored in MS. The objective of the study was to get insights into DTI-measurable pathological correlates of elevated serum GFAP in the normal appearing white matter (NAWM) of MS. METHODS A total of 62 MS patients with median age of 49.2 years were included in the study. Study patients underwent DTI-MRI and blood sampling for GFAP determination by single molecule array (Simoa). Mean fractional anisotropy (FA) and mean (MD), axial (AD) and radial (RD) diffusivities were calculated within the entire NAWM and six segmented NAWM regions. The associations between the DTI parameters and GFAP levels were analysed using Spearman correlation analysis and multiple regression model with sex and disease modifying treatment (no, 1st line or 2nd line) as adjustments. RESULTS Elevated serum GFAP levels correlated significantly with decreased FA values within the entire (ρ = -0.39, p = 0.03), frontal (ρ = -0.42, p = 0.02), temporal (ρ = -0.37; p = 0.04) and cingulate (ρ = -0.38, p = 0.034) NAWM, and increased MD and RD within the frontal NAWM (ρ = 0.36, p = 0.046 for both). Similarly, higher GFAP associated with lower FA in frontal and cingulate NAWM in the multiple regression model corrected for confounding variables (standardised regression coefficient β = -0.29, p = 0.045 and β = -0.33, p = 0.025). CONCLUSIONS Our results give evidence that increased serum GFAP levels associate with DTI-measurable micro-damage in the NAWM in MS. Our work supports the use of serum GFAP as a biomarker for MS pathology-related astrocytopathy and related diffuse white matter damage.
Collapse
Affiliation(s)
- Maija Saraste
- Turku PET Centre, Turku, Finland; Clinical Neurosciences, University of Turku, Turku, Finland; Neurocenter, Turku University Hospital, Turku, Finland.
| | | | - Markus Matilainen
- Turku PET Centre, Turku, Finland; Clinical Neurosciences, University of Turku, Turku, Finland; Neurocenter, Turku University Hospital, Turku, Finland.
| | - Marcus Sucksdorff
- Turku PET Centre, Turku, Finland; Clinical Neurosciences, University of Turku, Turku, Finland; Neurocenter, Turku University Hospital, Turku, Finland.
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland.
| | - David Leppert
- Neurological Clinic and Policlinic, University Hospital Basel, Basel, Switzerland.
| | - Laura Airas
- Turku PET Centre, Turku, Finland; Clinical Neurosciences, University of Turku, Turku, Finland; Neurocenter, Turku University Hospital, Turku, Finland.
| |
Collapse
|