1
|
Labib M, Thebault S, Booth RA, Brooks J, Rush C, MacLean H, Bose G, Freedman MS, Fadda G. The utility of serum neurofilament light chain in MOGAD: Current insights and future directions. Mult Scler Relat Disord 2025; 98:106410. [PMID: 40220725 DOI: 10.1016/j.msard.2025.106410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Serum neurofilament light chain (sNfL) has become an increasingly established biomarker for monitoring in multiple sclerosis (MS). Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is a demyelinating disorder distinct from MS in terms of pathophysiology and treatment options, also presenting with demyelinating attacks that can result in permanent disability. Given its unpredictable disease course, the need for biomarkers reflective of the risk for poor clinical recovery or relapsing course is pressing. The purpose of this review is to summarize the current knowledge on sNfL levels in people with MOGAD, assess their utility for clinical practice and gain insights for future research. METHODS Embase, MEDLINE, Scopus, and CINAHL databases were searched following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations. Keywords used in the search included: (myelin oligodendrocyte glycoprotein OR MOG OR MOGAD) AND (neurofilament* OR neuro filament* OR NfL OR sNfL). This initial search generated 195 reports, 23 of which were original research articles investigating NfL levels in MOGAD patients, therefore meeting our inclusion criteria. RESULTS 422 MOGAD patients were involved across all studies. Most studies revealed higher sNfL in MOGAD patients (n = 292) than in healthy controls (n = 3,172) with one study finding higher sNfL in MOGAD only during relapse. sNfL levels during attacks were similar when comparing MOGAD (n = 94) to MS (n = 256) and MOGAD (n = 149) to APQ4+ neuromyelitis optica spectrum disorder (APQ4+ NMOSD) (n = 214). MOGAD patients with brain lesions on magnetic resonance imaging (MRI) during a recent attack (n = 69 samples) had higher sNfL levels than patients without brain lesions (n = 78 samples). Median sNfL concentration was higher following clinical attacks (n = 69 samples) than in remission (n = 83 samples) in 3/5 studies. sNfL were higher at disease onset than subsequent attacks in 2 studies (n = 133 samples). Onset sNfL levels were not predictive of the likelihood of future relapse (relapsing: n = 15, monophasic: n = 18). A positive correlation was found between sNfL levels and attack severity assessed through various disability scales (n = 202), but not with the severity of acute or residual visual acuity (n = 45 eyes), or with residual retinal thickness among subjects with the optic neuritis (ON) phenotype (n = 11 eyes). The sGFAP/sNfL ratio showed utility in discriminating MOGAD from other autoimmune demyelinating diseases in two studies (MOGAD: n = 56, APQ4+ NMOSD: n = 66, MS: n = 31). DISCUSSION sNfL levels at presentation have limited utility in distinguishing MOGAD from other demyelinating disorders, but their combination with other biomarkers might improve their diagnostic utility. sNfL levels are higher in brain/spinal cord presentations than optic neuritis, correlating with clinical severity of these phenotypes but less so with the severity of visual outcome. Further studies should clarify the utility of sNfL as a biomarker for MOGAD, particularly in relation to long-term outcomes and imaging markers of central nervous system damage. Standardized sNfL testing parameters will improve study comparability and clinical application.
Collapse
Affiliation(s)
- Mark Labib
- Undergraduate Medical Education, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Simon Thebault
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Ronald A Booth
- Department of Pathology and Laboratory Medicine, University of Ottawa, The Ottawa Hospital and Eastern Ontario Regional Laboratory Association, Ottawa, ON, Canada.
| | - John Brooks
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - Carolina Rush
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - Heather MacLean
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - Gauruv Bose
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | - Giulia Fadda
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
2
|
Molitor TP, Hayashi G, Lin MY, Dunn CJ, Peterson NG, Poston RG, Kurnellas MP, Traver DA, Patel S, Akgungor Z, Leonardi V, Lewis C, Segales JS, Bennett DS, Truong AP, Dani M, Naphade S, Wong JK, McDermott AE, Kovalev SM, Ciaccio GL, Sadiq SA, Pei Z, Wood S, Rassoulpour A. Central TYK2 inhibition identifies TYK2 as a key neuroimmune modulator. Proc Natl Acad Sci U S A 2025; 122:e2422172122. [PMID: 40127268 PMCID: PMC12002270 DOI: 10.1073/pnas.2422172122] [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: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 03/26/2025] Open
Abstract
GWAS have identified tyrosine kinase 2 (TYK2) variants in multiple inflammatory disorders, specifically a protective hypomorphic TYK2 allele (P1104A) in multiple sclerosis (MS). Impaired TYK2 signaling within the central nervous system (CNS) may impart the protective effects of TYK2 P1104A allele in MS. We deployed brain-penetrant TYK2 inhibitors (cTYK2i) alongside the peripherally restricted TYK2 inhibitor (pTYK2i; BMS-986165) to untangle the contributions of central TYK2 inhibition in diverse models of neuroinflammation. While pTYK2i had little impact, cTYK2i reduced clinical score, lymphoid cell infiltration, and cytokines/chemokines in experimental autoimmune encephalomyelitis (EAE). Microglial activation was attenuated in cTYK2i-treated EAE spinal cords and circulating neurofilament light (NfL) was reduced in plasma and cerebral spinal fluid (CSF). Additionally, cTYK2i was protective in an antibody-mediated mouse model of primary progressive MS (PPMS). Finally, we demonstrate TYK2 inhibition has a robust impact on a unique subset of activated astrocytes termed Interferon-Responsive-Reactive-Astrocytes (IRRA). The data presented herein identify a key role for CNS TYK2 signaling in regulating neuroinflammation and solidify TYK2 as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Tyler P. Molitor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Genki Hayashi
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Mei-Yao Lin
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Carissa J. Dunn
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Robert G. Poston
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - David A. Traver
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Seona Patel
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Zeynep Akgungor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Colizel Lewis
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Dylan S. Bennett
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Anh P. Truong
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Manjari Dani
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Swati Naphade
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Jamie K. Wong
- Tisch MS Research Center of New York, New York, NY10019
| | | | | | | | - Saud A. Sadiq
- Tisch MS Research Center of New York, New York, NY10019
| | - Zhonghua Pei
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Stephen Wood
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | |
Collapse
|
3
|
Ali M, Wollenhaupt-Aguiar B, Sehmbi M, Minuzzi L, Bock NA, Frey BN. Peripheral neurofilament light chain and intracortical myelin in bipolar I disorder. J Affect Disord 2025; 374:184-190. [PMID: 39800065 DOI: 10.1016/j.jad.2025.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Neurofilament light chain (NfL) is a cytoskeletal protein that supports neuronal structure. Blood NfL levels are reported to be higher in diseases where myelin is damaged. Studies investigating intracortical myelin (ICM) in bipolar disorder (BD) have reported deficits in ICM maturation over age. This study investigated the association between ICM and peripheral blood NfL levels in BD. METHODS NfL was quantified using a high sensitivity ELISA kit in 72 BD and 71 healthy control (HC) participants. t-test was used to determine group difference in NfL levels between BD and HC, and ridge regression was performed to analyze NfL and ICM association in six brain regions that demonstrated ICM deficits including the dorsolateral motor cortex, dorsomedial motor cortex, dorsolateral premotor cortex, dorsomedial premotor cortex, caudal dorsolateral prefrontal cortex, caudal dorsomedial prefrontal cortex, and age in BD only. Regions within the occipital lobe and cingulum was also analyzed as control regions. RESULTS BD individuals had higher serum NfL concentration compared to HC (p = 0.001). The ridge regression analysis including the six brain regions and age explained 26 % of the variance in NfL concentration, while the occipital lobe and cingulum along with age explained only 7 % and 2 % of the variance, respectively. LIMITATIONS This was a cross-sectional correlational study so causation cannot be inferred. Also, this study focused on a limited number of brain regions previously associated with changes in ICM in BD. CONCLUSION This study corroborates previous research, which found increased NfL in CSF and blood in BD compared to HC. This demonstrates the potential utility of NfL as a marker of brain morphology deficits in BD.
Collapse
Affiliation(s)
- Mohammad Ali
- MiNDS Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, Canada
| | - Bianca Wollenhaupt-Aguiar
- Centre for Clinical Neurosciences, McMaster University, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada
| | - Manpreet Sehmbi
- MiNDS Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Luciano Minuzzi
- Centre for Clinical Neurosciences, McMaster University, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada
| | - Nicholas A Bock
- Department of Psychology, Neuroscience, and Behaviour, McMaster University, Hamilton, ON, Canada
| | - Benicio N Frey
- Centre for Clinical Neurosciences, McMaster University, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, Canada.
| |
Collapse
|
4
|
Samadzadeh S, Sleator RD. The role of Neurofilament light (NfL) and glial fibrillary acidic protein (GFAP) in MS and AQP4-NMOSD: Advancing clinical applications. eNeurologicalSci 2025; 38:100550. [PMID: 39866832 PMCID: PMC11762903 DOI: 10.1016/j.ensci.2025.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 01/28/2025] Open
Abstract
Fluid biomarkers such as Glial Fibrillary Acidic Protein (GFAP) and Neurofilament Light (NfL) play important roles in the diagnosis, monitoring, and evaluation of therapeutic responses in conditions such as Multiple Sclerosis (MS) and Aquaporin-4 Neuromyelitis Optica Spectrum Disorder (AQP4-NMOSD). These biomarkers offer key insights into the underlying pathophysiological mechanisms of these diseases, enabling effective follow-up and personalized treatment approaches, which are essential for improving patient outcomes. Herein, we synthesize the structural attributes, functional roles, and clinical significance of GFAP and NfL in the context of MS and AQP4-NMOSD. We explore the critical implications of these biomarkers in disease manifestation and progression, emphasizing the necessity to develop standardized methodologies and multicentric studies to confirm their clinical applicability.
Collapse
Affiliation(s)
- Sara Samadzadeh
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, Ireland
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospitals, Slagelse, Denmark
| | - Roy D. Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown, Cork, Ireland
| |
Collapse
|
5
|
Otsuki N, Kato T, Yokomura M, Urano M, Matsuo M, Kobayashi E, Haginoya K, Awano H, Takeshima Y, Saito T, Saito K. Analysis of SMN protein in umbilical cord blood and postnatal peripheral blood of neonates with SMA: a rationale for prompt treatment initiation to prevent SMA development. Orphanet J Rare Dis 2025; 20:91. [PMID: 40022154 PMCID: PMC11869478 DOI: 10.1186/s13023-025-03597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/08/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a severe genetic neuromuscular disease caused by insufficient functional survival motor neuron protein (SMN). The SMN expression level in the spinal cord is highest during the 2nd trimester of the foetal period. We previously reported the SMN spot analysis in peripheral blood using imaging flow cytometry (IFC) as a biomarker of functional SMN protein expression. In this study, we analysed neonatal cord blood, postnatal peripheral blood, and maternal peripheral blood in presymptomatic five infants whose sibling has type 1 SMA to estimate prenatal and postnatal SMN dynamics before the onset of severe SMA. RESULTS Data from 37 untreated patients with SMA showed that SMN-spot+ cells were significantly correlated with SMA clinical classification and the copy numbers of the SMN2 gene. The range of values for cord blood, converted from each SMN2 copy number statistics, was - 0.7 to + 2.0 standard deviation (SD) (0.1-24.0%) for SMN-spot+ cells in patients with SMA. Subsequent analyses of the peripheral blood of neonates ranged from - 0.8 to + 0.8 SD (0.4-15.2%). The analysis of each maternal blood, converted from carrier statistics, ranged from - 0.2 to + 2.4 SD (1.4-25.2%). A correlation was observed between the cord blood and maternal peripheral blood. CONCLUSIONS This study suggests that the status of the motor neuron pool in the spinal cord can be presumed by cord blood SMN-spot+ cells and that SMN protein depletion determines the timing of disease onset. As the SMN spot analysis values tended to decrease with time after birth, they may eventually lead to the development of SMA. Furthermore, a correlation was found between the SMN spot analysis values of neonatal cord blood and maternal blood, which predicts disease severity after birth. In other words, the SMN protein supplied from the mother to the foetus may suppress the development of SMA in the infant at birth, and depletion of the SMN protein may occur after birth, causing the infant to develop SMA. Our findings demonstrated the effectiveness of newborn screening and the potential of maternally mediated treatment strategies by providing a rationale for prompt treatment initiation in SMA.
Collapse
Affiliation(s)
- Noriko Otsuki
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Tamaki Kato
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Mamoru Yokomura
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Mari Urano
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Mari Matsuo
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Emiko Kobayashi
- Department of Pediatrics, Gifu Prefectural General Medical Center, 4-6-1 Noisshiki, Gifu City, Gifu, 500-8717, Japan
| | - Kazuhiro Haginoya
- Department of Pediatric Neurology, Miyagi Children's Hospital, 4-3-17 Ochiai, Aoba-ku, Sendai City, Miyagi, 989-3126, Japan
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe City, Hyogo, 650-0017, Japan
- Organization for Research Initiative and Promotion, Tottori University, 36-1 Nishi-cho, Yonago City, Tottori, 683-8503, Japan
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Toshio Saito
- Division of Child Neurology, Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Toyonaka City, Osaka, 560-8552, Japan
| | - Kayoko Saito
- Institute of Medical Genetics, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| |
Collapse
|
6
|
Sharma P, Giri A, Tripathi PN. Emerging Trends: Neurofilament Biomarkers in Precision Neurology. Neurochem Res 2024; 49:3208-3225. [PMID: 39347854 DOI: 10.1007/s11064-024-04244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024]
Abstract
Neurofilaments are structural proteins found in the cytoplasm of neurons, particularly in axons, providing structural support and stability to the axon. They consist of multiple subunits, including NF-H, NF-M, and NF-L, which form long filaments along the axon's length. Neurofilaments are crucial for maintaining the shape and integrity of neurons, promoting axonal transport, and regulating neuronal function. They are part of the intermediate filament (IF) family, which has approximately 70 tissue-specific genes. This diversity allows for a customizable cytoplasmic meshwork, adapting to the unique structural demands of different tissues and cell types. Neurofilament proteins show increased levels in both cerebrospinal fluid (CSF) and blood after neuroaxonal damage, indicating injury regardless of the underlying etiology. Precise measurement and long-term monitoring of damage are necessary for determining prognosis, assessing disease activity, tracking therapeutic responses, and creating treatments. These investigations contribute to our understanding of the importance of proper NF composition in fundamental neuronal processes and have implications for neurological disorders associated with NF abnormalities along with its alteration in different animal and human models. Here in this review, we have highlighted various neurological disorders such as Alzheimer's, Parkinson's, Huntington's, Dementia, and paved the way to use neurofilament as a marker in managing neurological disorders.
Collapse
Affiliation(s)
- Priti Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India
| | - Aditi Giri
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
| | - Prabhash Nath Tripathi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, India.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
7
|
Mrabet S, Sghaier I, Souissi A, Gharbi A, Abida Y, Kacem I, Gargouri-Berrechid A, Gouider R. Neurofilaments light chains as a diagnostic and predictive biomarker for Tunisian Multiple Sclerosis patients. Mult Scler Relat Disord 2024; 91:105901. [PMID: 39341199 DOI: 10.1016/j.msard.2024.105901] [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: 01/01/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Multiple Sclerosis (MS) course was shown to be more severe among North Africans compared to Caucasians. Validation of prognostic biomarkers of disease activity and severity is a priority in our practice. OBJECTIVE We aimed to investigate the association between baseline cerebrospinal fluid (CSF) and serum NfL (sNFL) levels and disease activity and disability accrual in a cohort of Tunisian patients with MS. METHODS A cross-sectional study was conducted, in the department of Neurology of Razi Hospital, including patients diagnosed with MS. Patient's data were retrieved from our local MS database. Blood and CSF sampling were performed at the first visit. sNFL levels were measured using the Enzyme-Linked Immuno-Sorbent Assay (ELISA) sandwich technique. RESULTS Three hundred MS patients were enrolled (sex-ratio= 3.05; mean age at MS onset=28.83 years+9.55, mean MS course = 10.21 years+8.96). MS phenotype was predominately relapsing (73%). CSF NfL levels were significantly correlated to the serum ones. NfL concentrations were significantly associated with MS activity (p = 0.012), disease progression (p = 0.001), and higher Multiple Sclerosis Severity Scores (MSSS) (p = 0.0017, r = 0.28). CONCLUSIONS These results support the value of NfL as a sensitive and clinically meaningful CSF and blood biomarker to evaluate MS activity and outcomes among Tunisian MS patients.
Collapse
Affiliation(s)
- Saloua Mrabet
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Ikram Sghaier
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Amira Souissi
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Alya Gharbi
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Youssef Abida
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Imen Kacem
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Amina Gargouri-Berrechid
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia
| | - Riadh Gouider
- Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, Tunis 1007, Tunisia; Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers, Manouba, Tunis 2010, Tunisia.
| |
Collapse
|
8
|
Cantero-Fortiz Y, Boada M. The role of inflammation in neurological disorders: a brief overview of multiple sclerosis, Alzheimer's, and Parkinson's disease'. Front Neurol 2024; 15:1439125. [PMID: 39539666 PMCID: PMC11558529 DOI: 10.3389/fneur.2024.1439125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroinflammation is a central feature in the pathophysiology of several neurodegenerative diseases, including MS, AD, and PD. This review aims to synthesize current research on the role of inflammation in these conditions, emphasizing the potential of inflammatory biomarkers for diagnosis and treatment. We highlight recent findings on the mechanisms of neuroinflammation, the utility of biomarkers in disease differentiation, and the implications for therapeutic strategies. Advances in understanding inflammatory pathways offer promising avenues for developing targeted interventions to improve patient outcomes. Future research should focus on validating these biomarkers in larger cohorts and integrating them into clinical practice to enhance diagnostic accuracy and therapeutic efficacy.
Collapse
Affiliation(s)
- Yahveth Cantero-Fortiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Lünemann JD, Sao Avilés A, Tintoré M, Midaglia L, Fissolo N, Gutiérrez L, Wiendl H, Montalban X, Comabella M. Cytomegalovirus immune responses are associated with lower serum NfL and disability accumulation risk at multiple sclerosis onset. Mult Scler 2024; 30:1445-1454. [PMID: 39246021 DOI: 10.1177/13524585241274571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
BACKGROUND Infection by cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) play a prognostic role in multiple sclerosis (MS). OBJECTIVES To explore whether humoral immune responses to HCMV and EBV at disease onset were associated with changes in serum and cerebrospinal fluid (CSF) levels of inflammatory and neurodegeneration biomarkers. METHODS Ninety-eight MS patients with a median follow-up of 20 years were included in the study. The levels of a panel of nine biomarkers were measured in serum (N = 60) and CSF (N = 61) samples of patients at the time of the first demyelinating event. RESULTS Immune responses to HCMV inversely correlated with serum neurofilament light chain (sNfL) levels (rho = -0.367; p = 0.039). sNfL levels were reduced in patients with high immune responses to HCMV (p = 0.006). Elevated sNfL levels were associated with higher risk of Expanded Disability Status Scale (EDSS) 3.0 (p = 0.016), 4.0 (p = 0.009) and 6.0 (p = 0.003), and with higher risk of developing secondary progressive MS (p = 0.003) and to receive treatment (p = 0.032). Serum soluble CD21 levels were increased in patients with high immune responses to EBV nuclear antigen 1 (p = 0.020). CONCLUSIONS High immune responses to HCMV are associated with limited disease progression and central nervous system (CNS) injury in MS patients. These findings reinforce the protective role of HCMV infection in MS.
Collapse
Affiliation(s)
- Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Augusto Sao Avilés
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Tintoré
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED)-ISCIII, Madrid, Spain
| | - Nicolás Fissolo
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED)-ISCIII, Madrid, Spain
| | - Lucía Gutiérrez
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Xavier Montalban
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED)-ISCIII, Madrid, Spain
| | - Manuel Comabella
- Unitat de Neuroimmunologia Clínica, Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED)-ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Højsgaard Chow H, Petersen ER, Olsson A, Hejgaard Laursen J, Bredahl Hansen M, Oturai AB, Soelberg Sørensen P, Bach Søndergaard H, Sellebjerg F. Age-corrected neurofilament light chain ratio decreases but does not predict relapse in highly active multiple sclerosis patients initiating natalizumab treatment. Mult Scler Relat Disord 2024; 88:105701. [PMID: 38889559 DOI: 10.1016/j.msard.2024.105701] [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: 12/06/2023] [Revised: 02/23/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Neurofilament light chain (NFL) is a biomarker for monitoring disease activity and treatment response in multiple sclerosis (MS). However, while most agree that NFL levels predict disease activity and worsening, the predictive value of NFL on future relapse risk remains uncertain. OBJECTIVE The primary aim was to evaluate the predictive value of age-corrected serum NFL (sNFL) ratio on relapse risk in highly active relapsing-remitting MS patients (RRMS) treated with natalizumab. A secondary aim was to investigate the predictive value of sNFL ratios for MRI activity. METHODS From January 1, 2006, to December 31, 2010, 355 patients initiated natalizumab treatment at the Danish Multiple Sclerosis Center. 305 patients were anti-natalizumab antibodies negative and had at least one blood sample available for sNFL analysis using single molecule array analysis at baseline, three, six, or 12 months. The patients were either treatment-naïve (n = 8), switching from interferon-β or glatiramer acetate (n = 253), or switching from mitoxantrone (n = 44). An age-corrected ratio was calculated for sNFL. Time to first relapse was calculated from baseline and after re-baseline at 90 days. Data were collected from baseline until the two-year follow-up or end of treatment and included disease duration, expanded disability status scale, previous treatments, relapses 12 months prior to natalizumab initiation, smoking intensity, body mass index, and body weight. In addition, the patients underwent annual MRI of the brain. RESULTS The sNFL ratio was increased in 173 of 287 samples (60.3 %) at baseline, in 119 of 246 samples (48.8 %) at month three, in 109 of 287 samples (38.0 %) at month six, and in 82 of 270 samples (30.4 %) at month 12. The sNFL ratio continuously declined over 12 months with significant decreases for every measuring timepoint: baseline vs. three months p = 3.0 × 10-6; three months vs. six months p = 3.2 × 10-5; six months vs. 12 months p = 0.002. Univariate Cox regression analysis showed that time to first relapse from 1) natalizumab initiation and from 2) re-baseline was associated with the number of relapses in the previous 12 months (hazard ratio 1.31 per relapse, 95 % CI = 1.2-1.5, p = 2.0 × 10-6; and 1.21 per relapse, 95 % CI = 1.1-1.4, p = 0.002, respectively). sNFL ratio at re-baseline was negatively associated with relapse risk (hazard ratio 0.82 per unit; 95 % CI = 0.7-1.0; p = 0.049). A multivariable Cox regression analysis of relapse risk from re-baseline showed that the number of relapses in the 12 months prior to natalizumab treatment (hazard ratio 1.29; 95 % CI = 1.1-1.5; p = 6.0 × 10-4) and smoking (hazard ratio 1.51 per 20 cigarettes per day; 95 % CI = 1.0-2.2; p = 0.030) were associated with increased risk of relapse; sNFL ratio was associated with a lower risk of relapse (hazard ratio = 0.736 per unit; 95 % CI = 0.6-0.9 p = 0.007). In univariate logistic regression analyses, the sNFL ratio at 12 months and values above the 75th and the 90th percentile predicted MRI activity in the following year (odds ratio [OR] = 2.0, 95 % CI = 1.2-3.6, p = 0.012; OR = 2.2, 95 % CI = 1.2-4.1, p = 0.014; and OR = 2.8, 95 % CI = 1.1-6.7, p = 0.026). CONCLUSION In this highly active RRMS cohort, high sNFL ratios reflected previous relapse activity and decreased after initiation of treatment but were not associated with increased relapse risk in the following two years. Pre-treatment relapses and smoking on treatment were predictors of relapse risk after re-baselining at 90 days. MRI activity in year two was predicted by sNFL ratios at month 12.
Collapse
Affiliation(s)
- Helene Højsgaard Chow
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| | - Eva Rosa Petersen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Anna Olsson
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Julie Hejgaard Laursen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Malene Bredahl Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Annette Bang Oturai
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Per Soelberg Sørensen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Helle Bach Søndergaard
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
11
|
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
|
12
|
Samadzadeh S, Adnan R, Berglova P, Barzegar M, Debrabant B, Roikjaer SG, Levy M, Petzold A, Palace J, Flanagan EP, Mariotto S, Skou ST, Froelich A, Lotan I, Messina S, Geraldes R, Asseyer S, Stiebel-Kalish H, Oertel FC, Shaygannejad V, Sahraian MA, Kim HJ, Bennett JL, Böttcher C, Zimmermann HG, Weinshenker BG, Paul F, Asgari N. Protocol of a prospective multicenter study on comorbidity impact on multiple sclerosis and antibody-mediated diseases of the central nervous system (COMMIT). Front Immunol 2024; 15:1380025. [PMID: 39021565 PMCID: PMC11253107 DOI: 10.3389/fimmu.2024.1380025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Comorbidities in patients with multiple sclerosis (MS) and antibody-mediated diseases of the central nervous system (CNS) including neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein (MOG)-antibody-associated disease (MOGAD) are common and may influence the course of their neurological disease. Comorbidity may contribute to neuronal injury and therefore limit recovery from attacks, accelerate disease progression, and increase disability. This study aims to explore the impact of comorbidity, particularly vascular comorbidity, and related risk factors on clinical and paraclinical parameters of MS, NMOSD and MOGAD. We propose COMMIT, a prospective multicenter study with longitudinal follow-up of patients with MS, NMOSD, and MOGAD, with or without comorbidities, as well as healthy subjects as controls. Subjects will be stratified by age, sex and ethnicity. In consecutive samples we will analyze levels of inflammation and neurodegeneration markers in both fluid and cellular compartments of the peripheral blood and cerebrospinal fluid (CSF) using multiple state-of-the-art technologies, including untargeted proteomics and targeted ultrasensitive ELISA assays and quantitative reverse transcription polymerase chain reaction (RT-qPCR) as well as high-dimensional single-cell technologies i.e., mass cytometry and single-cell RNA sequencing. Algorithm-based data analyses will be used to unravel the relationship between these markers, optical coherence tomography (OCT) and magnetic resonance imaging (MRI), and clinical outcomes including frequency and severity of relapses, long-term disability, and quality of life. The goal is to evaluate the impact of comorbidities on MS, NMOSD, and MOGAD which may lead to development of treatment approaches to improve outcomes of inflammatory demyelinating diseases of the CNS.
Collapse
Affiliation(s)
- Sara Samadzadeh
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
| | - Rafl Adnan
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
| | - Paulina Berglova
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
| | - Mahdi Barzegar
- Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Birgit Debrabant
- Department of Mathematics and Computer Science, Faculty of Natural Sciences, University of Southern Denmark, Odense, Denmark
| | - Stine Gundtoft Roikjaer
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- The Research and Implementation Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Region Zealand, Denmark
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Axel Petzold
- The National Hospital for Neurology and Neurosurgery, and Moorfields Eye Hospital and Queen Square University College London (UCL), Institute of Neurology, London, United Kingdom
- Department of Neurology, Amsterdam The University Medical Center Utrecht (UMC), Amsterdam, Netherlands
- Department of Ophthalmology, Amsterdam The University Medical Center Utrecht (UMC), Amsterdam, Netherlands
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Eoin P. Flanagan
- Department Neurology and Center for Multiple Sclerosis (MS), and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| | - Sara Mariotto
- Neurology Unit, Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| | - Soeren T. Skou
- The Research and Implementation Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Region Zealand, Denmark
- Center for Muscle and Joint Health, Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Anne Froelich
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Innovation and Research Centre for Multimorbidity, Slagelse Hospital, Slagelse, Denmark
- Section of General Practice, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Itay Lotan
- Department of Neurology and Neuroimmunology Unit, Rabin Medical Center, Petah Tikva, Israel
- Tel Aviv University Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Silvia Messina
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Ruth Geraldes
- Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, National Health Service Trust, Oxford, United Kingdom
| | - Susanna Asseyer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hadas Stiebel-Kalish
- Tel Aviv University Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Ophthalmology, Neuro-Ophthalmology Unit, Rabin Medical Center, Petah Tikva, Israel
- Eye Laboratory, Felsenstein Research Institute, Tel Aviv, Israel
| | - Frederike Cosima Oertel
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vahid Shaygannejad
- Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis (MS) Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Republic of Korea
| | - Jeffrey L. Bennett
- Department of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Chotima Böttcher
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hanna G. Zimmermann
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center Digital Future, Berlin, Germany
| | - Brian G. Weinshenker
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Friedemann Paul
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- NeuroCure Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nasrin Asgari
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- The Center for Neurological Research, Department of Neurology Slagelse Hospital, Slagelse, Denmark
- Open Patient data Explorative Network, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
13
|
Santorelli FM, McLoughlin HS, Wolter JM, Galatolo D, Synofzik M, Mengel D, Opal P. Standards of Fluid Biomarker Collection and Pre-analytical Processes in Humans and Mice: Recommendations by the Ataxia Global Initiative Working Group on Biomarkers. CEREBELLUM (LONDON, ENGLAND) 2024; 23:881-886. [PMID: 37243885 DOI: 10.1007/s12311-023-01561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/29/2023]
Abstract
The Ataxia Global Initiative (AGI) aims to serve as a platform to facilitate clinical trial readiness for the hereditary ataxias. Clinical trials for these diseases have been hampered by the lack of objective measures to study disease onset, progression, and treatment efficacy. While these issues are not unique to the genetic ataxias, the relative rarity of these diseases makes the need for such measures even more pressing to achieve statistical power in clinical trials. In this report, we have described the efforts of the AGI fluid biomarker working group (WG) in developing uniform protocols for biomarker sampling and storage, both for human and preclinical studies in mice. By reducing collection variability, we anticipate reduced noise in downstream biomarker analysis that will improve statistical power and minimize the necessary sample size. The emphasis has been on defining and standardizing the sampling and pre-analytical work-up of minimal set of biological samples, specifically blood plasma and serum, keeping in mind the need for harmonization of collection and storage that can be achieved with relatively limited cost and resources. An optional package is detailed for those centers that have the resources and commitment for additional biofluids/sample processing and storage. Finally, we have delineated similar standardized protocols for mice that will be important for preclinical studies in the field.
Collapse
Affiliation(s)
- Filippo M Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, Pisa, Italy.
| | | | - Justin M Wolter
- UNC Neuroscience Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Daniele Galatolo
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - David Mengel
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Puneet Opal
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
14
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Kizer K, Augusto DG, Tubati A, Gomez R, Fouassier C, Gerungan C, Caspar CM, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Sabatino JJ, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. An autoantibody signature predictive for multiple sclerosis. Nat Med 2024; 30:1300-1308. [PMID: 38641750 PMCID: PMC11980355 DOI: 10.1038/s41591-024-02938-3] [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: 04/18/2023] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. In this study, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster in approximately 10% of PwMS who share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active preclinical period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically or radiologically isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R Zamecnik
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gavin M Sowa
- University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ravi Dandekar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca D Bair
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kristen J Wade
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher M Bartley
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kerry Kizer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Danillo G Augusto
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Asritha Tubati
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Refujia Gomez
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Camille Fouassier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chloe Gerungan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Colette M Caspar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Alexander
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anne E Wapniarski
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rita P Loudermilk
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Erica L Eggers
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kelsey C Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nora Jabassini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sabrina A Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Nicholas R Ragan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Santaniello
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sergio E Baranzini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sabatino
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Riley M Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chu-Yueh Guo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey M Gelfand
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Richard Cuneo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - H-Christian von Büdingen
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jill A Hollenbach
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Ari J Green
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen L Hauser
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mitchell T Wallin
- Department of Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC, USA
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Michael R Wilson
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Ding EA, Kumar S. Neurofilament Biophysics: From Structure to Biomechanics. Mol Biol Cell 2024; 35:re1. [PMID: 38598299 PMCID: PMC11151108 DOI: 10.1091/mbc.e23-11-0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Neurofilaments (NFs) are multisubunit, neuron-specific intermediate filaments consisting of a 10-nm diameter filament "core" surrounded by a layer of long intrinsically disordered protein (IDP) "tails." NFs are thought to regulate axonal caliber during development and then stabilize the mature axon, with NF subunit misregulation, mutation, and aggregation featuring prominently in multiple neurological diseases. The field's understanding of NF structure, mechanics, and function has been deeply informed by a rich variety of biochemical, cell biological, and mouse genetic studies spanning more than four decades. These studies have contributed much to our collective understanding of NF function in axonal physiology and disease. In recent years, however, there has been a resurgence of interest in NF subunit proteins in two new contexts: as potential blood- and cerebrospinal fluid-based biomarkers of neuronal damage, and as model IDPs with intriguing properties. Here, we review established principles and more recent discoveries in NF structure and function. Where possible, we place these findings in the context of biophysics of NF assembly, interaction, and contributions to axonal mechanics.
Collapse
Affiliation(s)
- Erika A. Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
16
|
Desu HL, Sawicka KM, Wuerch E, Kitchin V, Quandt JA. A rapid review of differences in cerebrospinal neurofilament light levels in clinical subtypes of progressive multiple sclerosis. Front Neurol 2024; 15:1382468. [PMID: 38654736 PMCID: PMC11035744 DOI: 10.3389/fneur.2024.1382468] [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: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Background Multiple sclerosis (MS) is divided into three clinical phenotypes: relapsing-remitting MS (RRMS), secondary progressive MS (SPMS), and primary progressive MS (PPMS). It is unknown to what extent SPMS and PPMS pathophysiology share inflammatory or neurodegenerative pathological processes. Cerebrospinal (CSF) neurofilament light (NfL) has been broadly studied in different MS phenotypes and is a candidate biomarker for comparing MS subtypes. Research question Are CSF NfL levels different among clinical subtypes of progressive MS? Methods A search strategy identifying original research investigating fluid neurodegenerative biomarkers in progressive forms of MS between 2010 and 2022 was applied to Medline. Identified articles underwent title and abstract screen and full text review against pre-specified criteria. Data abstraction was limited to studies that measured NfL levels in the CSF. Reported statistical comparisons of NfL levels between clinical phenotypes were abstracted qualitatively. Results 18 studies that focused on investigating direct comparisons of CSF NfL from people with MS were included in the final report. We found NfL levels were typically reported to be higher in relapsing and progressive MS compared to healthy controls. Notably, higher NfL levels were not clearly associated with progressive MS subtypes when compared to relapsing MS, and there was no observed difference in NfL levels between PPMS and SPMS in articles that separately assessed these phenotypes. Conclusion CSF NfL levels distinguish individuals with MS from healthy controls but do not differentiate MS subtypes. Broad biological phenotyping is needed to overcome limitations of current clinical phenotyping and improve biomarker translatability to decision-making in the clinic.
Collapse
Affiliation(s)
- Haritha L. Desu
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| | - Katherine M. Sawicka
- Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Emily Wuerch
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada
| | - Vanessa Kitchin
- University of British Columbia Library, Vancouver, BC, Canada
| | - Jacqueline A. Quandt
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Kodosaki E, Watkins WJ, Loveless S, Kreft KL, Richards A, Anderson V, Hurler L, Robertson NP, Zelek WM, Tallantyre EC. Combination protein biomarkers predict multiple sclerosis diagnosis and outcomes. J Neuroinflammation 2024; 21:52. [PMID: 38368354 PMCID: PMC10874571 DOI: 10.1186/s12974-024-03036-4] [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: 10/09/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Establishing biomarkers to predict multiple sclerosis diagnosis and prognosis has been challenging using a single biomarker approach. We hypothesised that a combination of biomarkers would increase the accuracy of prediction models to differentiate multiple sclerosis from other neurological disorders and enhance prognostication for people with multiple sclerosis. We measured 24 fluid biomarkers in the blood and cerebrospinal fluid of 77 people with multiple sclerosis and 80 people with other neurological disorders, using ELISA or Single Molecule Array assays. Primary outcomes were multiple sclerosis versus any other diagnosis, time to first relapse, and time to disability milestone (Expanded Disability Status Scale 6), adjusted for age and sex. Multivariate prediction models were calculated using the area under the curve value for diagnostic prediction, and concordance statistics (the percentage of each pair of events that are correctly ordered in time for each of the Cox regression models) for prognostic predictions. Predictions using combinations of biomarkers were considerably better than single biomarker predictions. The combination of cerebrospinal fluid [chitinase-3-like-1 + TNF-receptor-1 + CD27] and serum [osteopontin + MCP-1] had an area under the curve of 0.97 for diagnosis of multiple sclerosis, compared to the best discriminative single marker in blood (osteopontin: area under the curve 0.84) and in cerebrospinal fluid (chitinase-3-like-1 area under the curve 0.84). Prediction for time to next relapse was optimal with a combination of cerebrospinal fluid[vitamin D binding protein + Factor I + C1inhibitor] + serum[Factor B + Interleukin-4 + C1inhibitor] (concordance 0.80), and time to Expanded Disability Status Scale 6 with cerebrospinal fluid [C9 + Neurofilament-light] + serum[chitinase-3-like-1 + CCL27 + vitamin D binding protein + C1inhibitor] (concordance 0.98). A combination of fluid biomarkers has a higher accuracy to differentiate multiple sclerosis from other neurological disorders and significantly improved the prediction of the development of sustained disability in multiple sclerosis. Serum models rivalled those of cerebrospinal fluid, holding promise for a non-invasive approach. The utility of our biomarker models can only be established by robust validation in different and varied cohorts.
Collapse
Affiliation(s)
- Eleftheria Kodosaki
- UK Dementia Research Institute at University College London, London, WC1E6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N3BG, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK
| | - W John Watkins
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Sam Loveless
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK
| | - Karim L Kreft
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| | - Aidan Richards
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK
| | - Valerie Anderson
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK
| | - Lisa Hurler
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, 1085, Hungary
| | - Neil P Robertson
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| | - Wioleta M Zelek
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Emma C Tallantyre
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF14 4XW, UK.
- Department of Neurology, University Hospital of Wales, Cardiff, UK.
| |
Collapse
|
18
|
Wilson D, Chan D, Chang L, Mathis R, Verberk I, Montalban X, Comabella M, Fissolo N, Bielekova B, Masvekar R, Chitnis T, Ziemssen T, Akgün K, Blennow K, Zetterberg H, Brück W, Giovannoni G, Gnanapavan S, Bittner S, Zipp F, Comi G, Furlan R, Lehmann S, Thebault S, Freedman M, Bar-Or A, Kramer M, Otto M, Halbgebauer S, Hrusovsky K, Plavina T, Khalil M, Piehl F, Wiendl H, Kappos L, Maceski A, Willemse E, Leppert D, Teunissen C, Kuhle J. Development and multi-center validation of a fully automated digital immunoassay for neurofilament light chain: toward a clinical blood test for neuronal injury. Clin Chem Lab Med 2024; 62:322-331. [PMID: 37702323 DOI: 10.1515/cclm-2023-0518] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVES Neurofilament light chain (NfL) has emerged as a promising biomarker for detecting and monitoring axonal injury. Until recently, NfL could only be reliably measured in cerebrospinal fluid, but digital single molecule array (Simoa) technology has enabled its precise measurement in blood samples where it is typically 50-100 times less abundant. We report development and multi-center validation of a novel fully automated digital immunoassay for NfL in serum for informing axonal injury status. METHODS A 45-min immunoassay for serum NfL was developed for use on an automated digital analyzer based on Simoa technology. The analytical performance (sensitivity, precision, reproducibility, linearity, sample type) was characterized and then cross validated across 17 laboratories in 10 countries. Analytical performance for clinical NfL measurement was examined in individual patients with relapsing remitting multiple sclerosis (RRMS) after 3 months of disease modifying treatment (DMT) with fingolimod. RESULTS The assay exhibited a lower limit of detection (LLoD) of 0.05 ng/L, a lower limit of quantification (LLoQ) of 0.8 ng/L, and between-laboratory imprecision <10 % across 17 validation sites. All tested samples had measurable NfL concentrations well above the LLoQ. In matched pre-post treatment samples, decreases in NfL were observed in 26/29 RRMS patients three months after DMT start, with significant decreases detected in a majority of patients. CONCLUSIONS The sensitivity characteristics and reproducible performance across laboratories combined with full automation make this assay suitable for clinical use for NfL assessment, monitoring in individual patients, and cross-comparisons of results across multiple sites.
Collapse
Affiliation(s)
| | | | | | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam, University Medical Centers, Amsterdam, The Netherlands
| | - Xavier Montalban
- Laboratori de Neuroinmunologia Clinica Centre d'Esclerosi Múltiple de Catalunya (Cemcat) Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Manuel Comabella
- Laboratori de Neuroinmunologia Clinica Centre d'Esclerosi Múltiple de Catalunya (Cemcat) Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Nicolas Fissolo
- Laboratori de Neuroinmunologia Clinica Centre d'Esclerosi Múltiple de Catalunya (Cemcat) Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Bibi Bielekova
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruturaj Masvekar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tanuja Chitnis
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tjalf Ziemssen
- MS Center Dresden, Center of Clinical Neuroscience, Department of Neurology, Dresden University of Technology, Dresden, Germany
| | - Katja Akgün
- MS Center Dresden, Center of Clinical Neuroscience, Department of Neurology, Dresden University of Technology, Dresden, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Wolfgang Brück
- Institute for Neuropathology at the University Medical Center, Göttingen, Germany
| | - Gavin Giovannoni
- Department of Neurology, Barts Health NHS Trust, The Royal London Hospital, E1 1FR, London, UK
| | - Sharmilee Gnanapavan
- Department of Neurology, Barts Health NHS Trust, The Royal London Hospital, E1 1FR, London, UK
| | - Stefan Bittner
- University Medical Center Mainz, Department of Neurology, Mainz, Germany
| | - Frauke Zipp
- University Medical Center Mainz, Department of Neurology, Mainz, Germany
| | - Giancarlo Comi
- Institute of Experimental Neurology, Division of Neuroscience, University Vita e Salute San Raffaele and IRCCS San Raffaele Hospital, Milan, Italy
| | - Roberto Furlan
- Institute of Experimental Neurology, Division of Neuroscience, University Vita e Salute San Raffaele and IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Simon Thebault
- University of Ottawa, Department of Medicine, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Mark Freedman
- University of Ottawa, Department of Medicine, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Amit Bar-Or
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Steffen Halbgebauer
- Department of Neurology, Ulm University Hospital, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE e.V.), Ulm, Germany
| | | | | | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Ludwig Kappos
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel, Departments of Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Aleksandra Maceski
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Eline Willemse
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - David Leppert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam, University Medical Centers, Amsterdam, The Netherlands
| | - Jens Kuhle
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Ghezzi A, Neuteboom RF. Neurofilament Light Chain in Adult and Pediatric Multiple Sclerosis: A Promising Biomarker to Better Characterize Disease Activity and Personalize MS Treatment. Neurol Ther 2023; 12:1867-1881. [PMID: 37682513 PMCID: PMC10630260 DOI: 10.1007/s40120-023-00535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
Many biological markers have been explored in multiple sclerosis (MS) to better quantify disease burden and better evaluate response to treatments, beyond clinical and MRI data. Among these, neurofilament light chain (Nf-L), although non-specific for this disease and found to be increased in other neurological conditions, has been shown to be the most promising biomarker for assessing axonal damage in MS, with a definite role in predicting the development of MS in patients at the first neurological episode suggestive of MS, and also in a preclinical phase. There is strong evidence that Nf-L levels are increased more in relapsing versus stable MS patients, and that they predict future disease evolution (relapses, progression, MRI measures of activity/progression) in MS patients, providing information on response to therapy, helping to anticipate clinical decisions in patients with an apparently stable evolution, and identifying patient non-responders to disease-modifying treatments. Moreover, Nf-L can contribute to the better understanding of the mechanisms of demyelination and axonal damage in adult and pediatric MS. A fundamental requirement for its clinical use is the accurate standardization of normal values, corrected for confounding factors, in particular age, sex, body mass index, and presence of comorbidities. In this review, a guide is provided to update clinicians on the use of Nf-L in clinical activity.
Collapse
Affiliation(s)
- Angelo Ghezzi
- Dipartimento di Scienze della Salute, Università Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100, Novara, Italy.
| | - R F Neuteboom
- Department of Neurology, ErasMS Center, Erasmus MC, PO Box 2040, 3000, Rotterdam, The Netherlands
| |
Collapse
|
20
|
dos Passos GR, Adoni T, Mendes MF, Sato DK. Reshaping neuroimmunology: diagnosis and treatment in the era of precision medicine. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:1125-1133. [PMID: 38157878 PMCID: PMC10756840 DOI: 10.1055/s-0043-1777752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024]
Abstract
Precision medicine has revolutionized the field of neuroimmunology, with innovative approaches that characterize diseases based on their biology, deeper understanding of the factors leading to heterogeneity within the same disease, development of targeted therapies, and strategies to tailor therapies to each patient. This review explores the impact of precision medicine on various neuroimmunological conditions, including multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), optic neuritis, autoimmune encephalitis, and immune-mediated neuropathies. We discuss advances in disease subtyping, recognition of novel entities, promising biomarkers, and the development of more selective monoclonal antibodies and cutting-edge synthetic cell-based immunotherapies in neuroimmunological disorders. In addition, we analyze the challenges related to affordability and equity in the implementation of these emerging technologies, especially in situations with limited resources.
Collapse
Affiliation(s)
- Giordani Rodrigues dos Passos
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina e Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
| | - Tarso Adoni
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, São Paulo SP, Brazil.
| | | | - Douglas Kazutoshi Sato
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina e Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brazil.
| |
Collapse
|
21
|
Jaromirska J, Kaczmarski P, Strzelecki D, Sochal M, Białasiewicz P, Gabryelska A. Shedding light on neurofilament involvement in cognitive decline in obstructive sleep apnea and its possible role as a biomarker. Front Psychiatry 2023; 14:1289367. [PMID: 38098628 PMCID: PMC10720906 DOI: 10.3389/fpsyt.2023.1289367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Obstructive sleep apnea is one of the most common sleep disorders with a high estimated global prevalence and a large number of associated comorbidities in general as well as specific neuropsychiatric complications such as cognitive impairment. The complex pathogenesis and effects of the disorder including chronic intermittent hypoxia and sleep fragmentation may lead to enhanced neuronal damage, thereby contributing to neuropsychiatric pathologies. Obstructive sleep apnea has been described as an independent risk factor for several neurodegenerative diseases, including Alzheimer's disease and all-cause dementia. The influence of obstructive sleep apnea on cognitive deficits is still a topic of recent debate, and several mechanisms, including neurodegeneration and depression-related cognitive dysfunction, underlying this correlation are taken into consideration. The differentiation between both pathomechanisms of cognitive impairment in obstructive sleep apnea is a complex clinical issue, requiring the use of multiple and costly diagnostic methods. The studies conducted on neuroprotection biomarkers, such as brain-derived neurotrophic factors and neurofilaments, are recently gaining ground in the topic of cognition assessment in obstructive sleep apnea patients. Neurofilaments as neuron-specific cytoskeletal proteins could be useful non-invasive indicators of brain conditions and neurodegeneration, which already are observed in many neurological diseases leading to cognitive deficits. Additionally, neurofilaments play an important role as a biomarker in other sleep disorders such as insomnia. Thus, this review summarizes the current knowledge on the involvement of neurofilaments in cognitive decline and neurodegeneration in obstructive sleep apnea patients as well as discusses its possible role as a biomarker of these changes.
Collapse
Affiliation(s)
- Julia Jaromirska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Piotr Kaczmarski
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
22
|
Zilinskaite N, Shukla RP, Baradoke A. Use of 3D Printing Techniques to Fabricate Implantable Microelectrodes for Electrochemical Detection of Biomarkers in the Early Diagnosis of Cardiovascular and Neurodegenerative Diseases. ACS MEASUREMENT SCIENCE AU 2023; 3:315-336. [PMID: 37868357 PMCID: PMC10588936 DOI: 10.1021/acsmeasuresciau.3c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
This Review provides a comprehensive overview of 3D printing techniques to fabricate implantable microelectrodes for the electrochemical detection of biomarkers in the early diagnosis of cardiovascular and neurodegenerative diseases. Early diagnosis of these diseases is crucial to improving patient outcomes and reducing healthcare systems' burden. Biomarkers serve as measurable indicators of these diseases, and implantable microelectrodes offer a promising tool for their electrochemical detection. Here, we discuss various 3D printing techniques, including stereolithography (SLA), digital light processing (DLP), fused deposition modeling (FDM), selective laser sintering (SLS), and two-photon polymerization (2PP), highlighting their advantages and limitations in microelectrode fabrication. We also explore the materials used in constructing implantable microelectrodes, emphasizing their biocompatibility and biodegradation properties. The principles of electrochemical detection and the types of sensors utilized are examined, with a focus on their applications in detecting biomarkers for cardiovascular and neurodegenerative diseases. Finally, we address the current challenges and future perspectives in the field of 3D-printed implantable microelectrodes, emphasizing their potential for improving early diagnosis and personalized treatment strategies.
Collapse
Affiliation(s)
- Nemira Zilinskaite
- Wellcome/Cancer
Research UK Gurdon Institute, Henry Wellcome Building of Cancer and
Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K.
- Faculty
of Medicine, University of Vilnius, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
| | - Rajendra P. Shukla
- BIOS
Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck
Center for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Ausra Baradoke
- Wellcome/Cancer
Research UK Gurdon Institute, Henry Wellcome Building of Cancer and
Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K.
- Faculty
of Medicine, University of Vilnius, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
- BIOS
Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck
Center for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- Center for
Physical Sciences and Technology, Savanoriu 231, LT-02300 Vilnius, Lithuania
| |
Collapse
|
23
|
Duran-Castells C, Prats A, Oriol-Tordera B, Llano A, Galvez C, Martinez-Picado J, Ballana E, Garcia-Vidal E, Clotet B, Muñoz-Moreno JA, Hanke T, Moltó J, Mothe B, Brander C, Ruiz-Riol M. Plasma proteomic profiling identifies CD33 as a marker of HIV control in natural infection and after therapeutic vaccination. EBioMedicine 2023; 95:104732. [PMID: 37506557 PMCID: PMC10410179 DOI: 10.1016/j.ebiom.2023.104732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Biomarkers predicting the outcome of HIV-1 virus control in natural infection and after therapeutic interventions in HIV-1 cure trials remain poorly defined. The BCN02 trial (NCT02616874), combined a T-cell vaccine with romidepsin (RMD), a cancer-drug that was used to promote HIV-1 latency reversal and which has also been shown to have beneficial effects on neurofunction. We conducted longitudinal plasma proteomics analyses in trial participants to define biomarkers associated with virus control during monitored antiretroviral pause (MAP) and to identify novel therapeutic targets that can improve future cure strategies. METHODS BCN02 was a phase I, open-label, single-arm clinical trial in early-treated, HIV infected individuals. Longitudinal plasma proteomes were analyzed in 11 BCN02 participants, including 8 participants that showed a rapid HIV-1 plasma rebound during a monitored antiretroviral pause (MAP-NC, 'non-controllers') and 3 that remained off ART with sustained plasma viremia <2000 copies/ml (MAP-C, 'controllers'). Inflammatory and neurological proteomes in plasma were evaluated and integration data analysis (viral and neurocognitive parameters) was performed. Validation studies were conducted in a cohort of untreated HIV-1+ individuals (n = 96) and in vitro viral replication assays using an anti-CD33 antibody were used for functional validation. FINDINGS Inflammatory plasma proteomes in BCN02 participants showed marked longitudinal alterations. Strong proteome differences were also observed between MAP-C and MAP-NC, including in baseline timepoints. CD33/Siglec-3 was the unique plasma marker with the ability to discriminate between MAPC-C and MAP-NC at all study timepoints and showed positive correlations with viral parameters. Analyses in an untreated cohort of PLWH confirmed the positive correlation between viral parameters and CD33 plasma levels, as well as PBMC gene expression. Finally, adding an anti-CD33 antibody to in vitro virus cultures significantly reduced HIV-1 replication and proviral levels in T cells and macrophages. INTERPRETATION This study indicates that CD33/Siglec-3 may serve as a predictor of HIV-1 control and as potential therapeutic tool to improve future cure strategies. FUNDING Spanish Science and Innovation Ministry (SAF2017-89726-R and PID2020-119710RB-I00), NIH (P01-AI131568), European Commission (GA101057548) and a Grifols research agreement.
Collapse
Affiliation(s)
- Clara Duran-Castells
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Universitat Autònoma de Barcelona, Spain
| | - Anna Prats
- Fight Infections Foundation and Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Bruna Oriol-Tordera
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Universitat Autònoma de Barcelona, Spain
| | - Anuska Llano
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Cristina Galvez
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain; Catalan Institution for Research Advanced Studies (ICREA), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Ester Ballana
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Edurne Garcia-Vidal
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Fight Infections Foundation and Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Jose A Muñoz-Moreno
- Fight Infections Foundation and Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Faculty of Psychology and Education Sciences, Universitat Oberta de Catalunya (UOC), Barcelona, Spain
| | - Thomas Hanke
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - José Moltó
- Fight Infections Foundation and Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Fight Infections Foundation and Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Universitat Autònoma de Barcelona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain; Catalan Institution for Research Advanced Studies (ICREA), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute Badalona, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| |
Collapse
|
24
|
Pryce G, Sisay S, Giovannoni G, Selwood DL, Baker D. Neuroprotection in an Experimental Model of Multiple Sclerosis via Opening of Big Conductance, Calcium-Activated Potassium Channels. Pharmaceuticals (Basel) 2023; 16:972. [PMID: 37513884 PMCID: PMC10383993 DOI: 10.3390/ph16070972] [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: 05/23/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Big conductance calcium-activated (BK) channel openers can inhibit pathologically driven neural hyperactivity to control symptoms via hyperpolarizing signals to limit neural excitability. We hypothesized that BK channel openers would be neuroprotective during neuroinflammatory, autoimmune disease. The neurodegenerative disease was induced in a mouse experimental autoimmune encephalomyelitis model with translational value to detect neuroprotection in multiple sclerosis. Following the treatment with the BK channel openers, BMS-204253 and VSN16R, neuroprotection was assessed using subjective and objective clinical outcomes and by quantitating spinal nerve content. Treatment with BMS-204253 and VSN16R did not inhibit the development of relapsing autoimmunity, consistent with minimal channel expression via immune cells, nor did it change leukocyte levels in rodents or humans. However, it inhibited the accumulation of nerve loss and disability as a consequence of autoimmunity. Therefore, in addition to symptom control, BK channel openers have the potential to save nerves from excitotoxic damage and could be useful as either stand-alone neuroprotective agents or as add-ons to current disease-modifying treatments that block relapsing MS but do not have any direct neuroprotective activity.
Collapse
Affiliation(s)
- Gareth Pryce
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Sofia Sisay
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Gavin Giovannoni
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - David L Selwood
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - David Baker
- BartsMS, The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
25
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Tubati A, Gomez R, Fouassier C, Gerungan C, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. A Predictive Autoantibody Signature in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.01.23288943. [PMID: 37205595 PMCID: PMC10187343 DOI: 10.1101/2023.05.01.23288943] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. Here, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster of PwMS that share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active prodromal period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid (CSF) and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically- or radiologically-isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R. Zamecnik
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Gavin M. Sowa
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Ravi Dandekar
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rebecca D. Bair
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kristen J. Wade
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Christopher M. Bartley
- UCSF Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Asritha Tubati
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Refujia Gomez
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Camille Fouassier
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chloe Gerungan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jessica Alexander
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne E. Wapniarski
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rita P. Loudermilk
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Erica L. Eggers
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kelsey C. Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sabrina A. Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Nicholas R. Ragan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Adam Santaniello
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Roland G. Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sergio E. Baranzini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Scott S. Zamvil
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Riley M. Bove
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chu-Yueh Guo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jeffrey M. Gelfand
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Richard Cuneo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - H.-Christian von Büdingen
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jorge R. Oksenberg
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce AC Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jill A. Hollenbach
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA USA
| | - Ari J. Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Stephen L. Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Mitchell T. Wallin
- Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Michael R. Wilson
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
26
|
Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. NATURE AGING 2023; 3:506-519. [PMID: 37202517 PMCID: PMC10979350 DOI: 10.1038/s43587-023-00403-3] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|