1
|
Agah E, Mojtabavi H, Behkar A, Heidari A, Ajdari A, Shaka Z, Mousavi SV, Firoozeh N, Tafakhori A, Rezaei N. CSF and blood levels of Neurofilaments, T-Tau, P-Tau, and Abeta-42 in amyotrophic lateral sclerosis: a systematic review and meta-analysis. J Transl Med 2024; 22:953. [PMID: 39434139 PMCID: PMC11492992 DOI: 10.1186/s12967-024-05767-7] [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: 07/22/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
Recent literature suggests that markers of neuroaxonal damage, such as neurofilaments and tau protein, might serve as potential biomarkers for ALS. We conducted this systematic review and meta-analysis study to compare cerebrospinal fluid (CSF) and blood levels of total tau (t-tau), phosphorylated tau (p-tau), amyloid beta peptide 42 (Abeta-42), and neurofilaments in ALS patients and controls. A systematic search of Cochrane Library, PubMed, Embase, and ISI Web of Science was conducted on March 18, 2022, and updated on January 26, 2023. Observational studies that compared the concentrations of neurofilament light chain (NfL), neurofilament heavy chain (NFH), t-tau, p-tau, or Abeta-42 in CSF or peripheral blood of ALS patients and controls were included. Data from relevant studies were independently extracted and screened for quality using a standard tool, by at least two authors. Meta-analysis was conducted when a minimum of 3 studies reported the same biomarker within the same biofluid. A total of 100 studies were eligible for at least one meta-analysis. CSF and blood levels of NfL (standardized mean difference (SMD) [95% CI]; CSF: 1.46 [1.25-1.68]; blood: 1.35 [1.09-1.60]) and NFH (CSF: 1.32 [1.13-1.50], blood: 0.90 [0.58-1.22]) were significantly higher in ALS patients compared with controls. The pooled differences between ALS patients and controls were not significant for CSF t-tau, blood t-tau, and CSF Abeta-42, but CSF p-tau was lower in ALS patients (-0.27 [-0.47- -0.07]). Significantly decreased p-tau/t-tau ratios were found in ALS patients compared with controls (-0.84 [-1.16- -0.53]). Heterogeneity was considerable in most of our meta-analyses. CSF and blood neurofilament levels, as well as the CSF p-tau/t-tau ratio, might be potential candidates for improving ALS diagnosis. Further research is warranted to better understand the underlying mechanisms and the clinical implications of these biomarker alterations.
Collapse
Affiliation(s)
- Elmira Agah
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Helia Mojtabavi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- National Center for Adaptive Neurotechnologies (NCAN), Albany, NY, USA
| | - Atefeh Behkar
- Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Heidari
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies (RCID), Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atra Ajdari
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zoha Shaka
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Vahid Mousavi
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Firoozeh
- Department of Radiology, Harborview Medical Center, University of Washington, Seattle, WA, USA
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nima Rezaei
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
2
|
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
|
3
|
Shahim P, Norato G, Sinaii N, Zetterberg H, Blennow K, Chan L, Grunseich C. Neurofilaments in Sporadic and Familial Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. Genes (Basel) 2024; 15:496. [PMID: 38674431 PMCID: PMC11050235 DOI: 10.3390/genes15040496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Neurofilament proteins have been implicated to be altered in amyotrophic lateral sclerosis (ALS). The objectives of this study were to assess the diagnostic and prognostic utility of neurofilaments in ALS. METHODS Studies were conducted in electronic databases (PubMed/MEDLINE, Embase, Web of Science, and Cochrane CENTRAL) from inception to 17 August 2023, and investigated neurofilament light (NfL) or phosphorylated neurofilament heavy chain (pNfH) in ALS. The study design, enrolment criteria, neurofilament concentrations, test accuracy, relationship between neurofilaments in cerebrospinal fluid (CSF) and blood, and clinical outcome were recorded. The protocol was registered with PROSPERO, CRD42022376939. RESULTS Sixty studies with 8801 participants were included. Both NfL and pNfH measured in CSF showed high sensitivity and specificity in distinguishing ALS from disease mimics. Both NfL and pNfH measured in CSF correlated with their corresponding levels in blood (plasma or serum); however, there were stronger correlations between CSF NfL and blood NfL. NfL measured in blood exhibited high sensitivity and specificity in distinguishing ALS from controls. Both higher levels of NfL and pNfH either measured in blood or CSF were correlated with more severe symptoms as assessed by the ALS Functional Rating Scale Revised score and with a faster disease progression rate; however, only blood NfL levels were associated with shorter survival. DISCUSSION Both NfL and pNfH measured in CSF or blood show high diagnostic utility and association with ALS functional scores and disease progression, while CSF NfL correlates strongly with blood (either plasma or serum) and is also associated with survival, supporting its use in clinical diagnostics and prognosis. Future work must be conducted in a prospective manner with standardized bio-specimen collection methods and analytical platforms, further improvement in immunoassays for quantification of pNfH in blood, and the identification of cut-offs across the ALS spectrum and controls.
Collapse
Affiliation(s)
- Pashtun Shahim
- Rehabilitation Medicine Department, National Institutes of Health (NIH) Clinical Center, Bethesda, MD 20892, USA;
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
- Department of Neurology, MedStar Georgetown University Hospital, Washington, DC 20007, USA
- The Military Traumatic Brain Injury Initiative (MTBI2), Bethesda, MD 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Gina Norato
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, NIH, Bethesda, MD 20892, USA;
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Molndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahglrenska University Hospital, 431 41 Molndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 518172, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Molndal, Sweden; (H.Z.); (K.B.)
- Clinical Neurochemistry Laboratory, Sahglrenska University Hospital, 431 41 Molndal, Sweden
| | - Leighton Chan
- Rehabilitation Medicine Department, National Institutes of Health (NIH) Clinical Center, Bethesda, MD 20892, USA;
| | - Christopher Grunseich
- National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA; (G.N.); (C.G.)
| |
Collapse
|
4
|
Vrillon A, Ashton NJ, Karikari TK, Götze K, Cognat E, Dumurgier J, Lilamand M, Zetterberg H, Blennow K, Paquet C. Comparison of CSF and plasma NfL and pNfH for Alzheimer's disease diagnosis: a memory clinic study. J Neurol 2024; 271:1297-1310. [PMID: 37950758 DOI: 10.1007/s00415-023-12066-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/13/2023]
Abstract
Plasma neurofilament light chain (NfL) is a promising biomarker of axonal damage for the diagnosis of neurodegenerative diseases. Phosphorylated neurofilament heavy chain (pNfH) has demonstrated its value in motor neuron diseases diagnosis, but has less been explored for dementia diagnosis. In a cross-sectional study, we compared cerebrospinal fluid (CSF) and plasma NfL and pNfH levels in n = 188 patients from Lariboisière Hospital, Paris, France, including AD patients at mild cognitive impairment stage (AD-MCI, n = 36) and dementia stage (n = 64), non-AD MCI (n = 38), non-AD dementia (n = 28) patients and control subjects (n = 22). Plasma NfL, plasma and CSF pNfH levels were measured using Simoa and CSF NfL using ELISA. The correlation between CSF and plasma levels was stronger for NfL than pNfH (rho = 0.77 and rho = 0.52, respectively). All neurofilament markers were increased in AD-MCI, AD dementia and non-AD dementia groups compared with controls. CSF NfL, CSF pNfH and plasma NfL showed high performance to discriminate AD at both MCI and dementia stages from control subjects [AUC (area under the curve) = 0.82-0.91]. Plasma pNfH displayed overall lower AUCs for discrimination between groups compared with CSF pNfH. Neurofilament markers showed similar moderate association with cognition. NfL levels displayed significant association with mediotemporal lobe atrophy and white matter lesions in the AD group. Our results suggest that CSF NfL and pNfH as well as plasma NfL levels display equivalent performance in both positive and differential AD diagnosis in memory clinic settings. In contrast to motoneuron disorders, plasma pNfH did not demonstrate added value as compared with plasma NfL.
Collapse
Affiliation(s)
- Agathe Vrillon
- Cognitive Neurology Center, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France.
- INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France.
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Maurice Wohl Institute Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Götze
- INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Emmanuel Cognat
- Cognitive Neurology Center, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
- INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Julien Dumurgier
- Cognitive Neurology Center, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Matthieu Lilamand
- Cognitive Neurology Center, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Maurice Wohl Institute Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Claire Paquet
- Cognitive Neurology Center, Lariboisière Fernand Widal Hospital, Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France
- INSERM U1144, Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| |
Collapse
|
5
|
Bellanti R, Keddie S, Lunn MP, Rinaldi S. Ultrasensitive assay technology and fluid biomarkers for the evaluation of peripheral nerve disease. J Neurol Neurosurg Psychiatry 2024; 95:114-124. [PMID: 37821222 DOI: 10.1136/jnnp-2023-332031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 10/13/2023]
Abstract
The field of biomarker discovery is rapidly expanding. The introduction of ultrasensitive immunoassays and the growing precision of genetic technologies are poised to revolutionise the assessment and monitoring of many diseases. Given the difficulties in imaging and tissue diagnosis, there is mounting interest in serum and cerebrospinal fluid biomarkers of peripheral neuropathy. Realised and potential fluid biomarkers of peripheral nerve disease include neuronal biomarkers of axonal degeneration, glial biomarkers for peripheral demyelinating disorders, immunopathogenic biomarkers (such as the presence and titre of antibodies or the levels of cytokines) and genetic biomarkers. Several are already starting to inform clinical practice, whereas others remain under evaluation as potential indicators of disease activity and treatment response. As more biomarkers become available for clinical use, it has become increasingly difficult for clinicians and researchers to keep up-to-date with the most recent discovery and interpretation. In this review, we aim to inform practising neurologists, neuroscientists and other clinicians about recent advances in fluid biomarker technology, with a focus on single molecule arrays (Simoa), chemiluminescent enzyme immunoassays (CLEIA), electrochemiluminescence (ECL), proximity extension assays (PEA), and microfluidic technology. We discuss established and emerging fluid biomarkers of peripheral neuropathy, their clinical applications, limitations and potential future developments.
Collapse
Affiliation(s)
- Roberto Bellanti
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Stephen Keddie
- Department of Neuromuscular Diseases, The Royal London Hospital, London, UK
| | - Michael P Lunn
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
- Department of Neuroinflammation, National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Mimic S, Aru B, Pehlivanoğlu C, Sleiman H, Andjus PR, Yanıkkaya Demirel G. Immunology of amyotrophic lateral sclerosis - role of the innate and adaptive immunity. Front Neurosci 2023; 17:1277399. [PMID: 38105925 PMCID: PMC10723830 DOI: 10.3389/fnins.2023.1277399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
This review aims to summarize the latest evidence about the role of innate and adaptive immunity in Amyotrophic Lateral Sclerosis (ALS). ALS is a devastating neurodegenerative disease affecting upper and lower motor neurons, which involves essential cells of the immune system that play a basic role in innate or adaptive immunity, that can be neurotoxic or neuroprotective for neurons. However, distinguishing between the sole neurotoxic or neuroprotective function of certain cells such as astrocytes can be challenging due to intricate nature of these cells, the complexity of the microenvironment and the contextual factors. In this review, in regard to innate immunity we focus on the involvement of monocytes/macrophages, microglia, the complement, NK cells, neutrophils, mast cells, and astrocytes, while regarding adaptive immunity, in addition to humoral immunity the most important features and roles of T and B cells are highlighted, specifically different subsets of CD4+ as well as CD8+ T cells. The role of autoantibodies and cytokines is also discussed in distinct sections of this review.
Collapse
Affiliation(s)
- Stefan Mimic
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Hadi Sleiman
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
7
|
Matschke V, Kürten K, Gude AC, Christian Epplen A, Stein J, Theiss C. Dysregulated expression and distribution of Kif5α in neurites of wobbler motor neurons. Neural Regen Res 2023. [PMID: 35799535 PMCID: PMC9241431 DOI: 10.4103/1673-5374.343883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Impaired axonal transport has been observed in patients with amyotrophic lateral sclerosis (ALS) and in animal models, suggesting that transport proteins likely play a critical role in the pathological mechanism of ALS. Dysregulation of Kinesin-family-member 5α (Kif5α), a neuron-specific isoform of heavy chain kinesin family, has been described in several neurological disorders, in humans and animal models, including ALS. In this study, we determined Kif5α expression by gene sequencing, quantitative reverse transcription-polymerase chain reaction, and western blot assay in the cervical spinal cord of wobbler mice and immunofluorescence staining in dissociated cultures of the ventral horn. Further, we observed the distribution of Kif5α and mitochondria along motor neuronal branches by confocal imaging. Our results showed that Kif5α expression was greatly dysregulated in wobbler mice, which resulted in altered distribution of Kif5α along motor neuronal branches with an abnormal mitochondrial distribution. Thus, our results indicate that dysregulation of Kif5 and therefore abnormal transport in motor neuronal branches in this ALS model could be causative for several pathological findings at the cellular level, like misallocation of cytoskeletal proteins or organelles like mitochondria.
Collapse
|
8
|
Waury K, Willemse EAJ, Vanmechelen E, Zetterberg H, Teunissen CE, Abeln S. Bioinformatics tools and data resources for assay development of fluid protein biomarkers. Biomark Res 2022; 10:83. [DOI: 10.1186/s40364-022-00425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractFluid protein biomarkers are important tools in clinical research and health care to support diagnosis and to monitor patients. Especially within the field of dementia, novel biomarkers could address the current challenges of providing an early diagnosis and of selecting trial participants. While the great potential of fluid biomarkers is recognized, their implementation in routine clinical use has been slow. One major obstacle is the often unsuccessful translation of biomarker candidates from explorative high-throughput techniques to sensitive antibody-based immunoassays. In this review, we propose the incorporation of bioinformatics into the workflow of novel immunoassay development to overcome this bottleneck and thus facilitate the development of novel biomarkers towards clinical laboratory practice. Due to the rapid progress within the field of bioinformatics many freely available and easy-to-use tools and data resources exist which can aid the researcher at various stages. Current prediction methods and databases can support the selection of suitable biomarker candidates, as well as the choice of appropriate commercial affinity reagents. Additionally, we examine methods that can determine or predict the epitope - an antibody’s binding region on its antigen - and can help to make an informed choice on the immunogenic peptide used for novel antibody production. Selected use cases for biomarker candidates help illustrate the application and interpretation of the introduced tools.
Collapse
|
9
|
Petzold A. The 2022 Lady Estelle Wolfson lectureship on neurofilaments. J Neurochem 2022; 163:179-219. [PMID: 35950263 PMCID: PMC9826399 DOI: 10.1111/jnc.15682] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 01/11/2023]
Abstract
Neurofilament proteins (Nf) have been validated and established as a reliable body fluid biomarker for neurodegenerative pathology. This review covers seven Nf isoforms, Nf light (NfL), two splicing variants of Nf medium (NfM), two splicing variants of Nf heavy (NfH),α -internexin (INA) and peripherin (PRPH). The genetic and epigenetic aspects of Nf are discussed as relevant for neurodegenerative diseases and oncology. The comprehensive list of mutations for all Nf isoforms covers Amyotrophic Lateral Sclerosis, Charcot-Marie Tooth disease, Spinal muscular atrophy, Parkinson Disease and Lewy Body Dementia. Next, emphasis is given to the expanding field of post-translational modifications (PTM) of the Nf amino acid residues. Protein structural aspects are reviewed alongside PTMs causing neurodegenerative pathology and human autoimmunity. Molecular visualisations of NF PTMs, assembly and stoichiometry make use of Alphafold2 modelling. The implications for Nf function on the cellular level and axonal transport are discussed. Neurofilament aggregate formation and proteolytic breakdown are reviewed as relevant for biomarker tests and disease. Likewise, Nf stoichiometry is reviewed with regard to in vitro experiments and as a compensatory mechanism in neurodegeneration. The review of Nf across a spectrum of 87 diseases from all parts of medicine is followed by a critical appraisal of 33 meta-analyses on Nf body fluid levels. The review concludes with considerations for clinical trial design and an outlook for future research.
Collapse
Affiliation(s)
- Axel Petzold
- Department of NeurodegenerationQueen Square Insitute of Neurology, UCLLondonUK
| |
Collapse
|
10
|
Sturmey E, Malaspina A. Blood biomarkers in ALS: challenges, applications and novel frontiers. Acta Neurol Scand 2022; 146:375-388. [PMID: 36156207 PMCID: PMC9828487 DOI: 10.1111/ane.13698] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/12/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease among adults. With diagnosis reached relatively late into the disease process, extensive motor cell loss narrows the window for therapeutic opportunities. Clinical heterogeneity in ALS and the lack of disease-specific biomarkers have so far led to large-sized clinical trials with long follow-up needed to define clinical outcomes. In advanced ALS patients, there is presently limited scope to use imaging or invasive cerebrospinal fluid (CSF) collection as a source of disease biomarkers. The development of more patient-friendly and accessible blood biomarker assays is hampered by analytical hurdles like the matrix effect of blood components. However, blood also provides the opportunity to identify disease-specific adaptive changes of the stoichiometry and conformation of target proteins and the endogenous immunological response to low-abundance brain peptides, such as neurofilaments (Nf). Among those biomarkers under investigation in ALS, the change in concentration before or after diagnosis of Nf has been shown to aid prognostication and to allow the a priori stratification of ALS patients into smaller sized and clinically more homogeneous cohorts, supporting more affordable clinical trials. Here, we discuss the technical hurdles affecting reproducible and sensitive biomarker measurement in blood. We also summarize the state of the art of non-CSF biomarkers in the study of prognosis, disease progression, and treatment response. We will then address the potential as disease-specific biomarkers of the newly discovered cryptic peptides which are formed down-stream of TDP-43 loss of function, the hallmark of ALS pathobiology.
Collapse
Affiliation(s)
- Ellie Sturmey
- Centre of Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK
| | - Andrea Malaspina
- Centre of Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK.,Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
11
|
Zecca C, Dell'Abate MT, Pasculli G, Capozzo R, Barone R, Arima S, Pollice A, Brescia V, Tortelli R, Logroscino G. Role of plasma phosphorylated neurofilament heavy chain (pNfH) in amyotrophic lateral sclerosis. J Cell Mol Med 2022; 26:3608-3615. [PMID: 35715961 PMCID: PMC9258711 DOI: 10.1111/jcmm.17232] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/26/2022] Open
Abstract
The phosphorylated neurofilament heavy chain (pNfH) is a promising biomarker in amyotrophic lateral sclerosis (ALS). We examined plasma pNfH concentrations in order to corroborate its role as a diagnostic and prognostic biomarker in ALS. Incident ALS cases enrolled in a population‐based registry were retrospectively selected and matched by sex and age with a cohort of healthy volunteers. Plasma pNfH levels were measured by an ELISA kit and correlated with clinical parameters. Discrimination ability of pNfH was tested using receiving operating characteristic (ROC) curves. Kaplan–Meier (KM) analysis and Cox proportional hazard models were used for survival analysis. Plasma pNfH was significantly higher in patients compared to controls. An optimal cut‐off of 39.74 pg/ml discriminated cases from controls with an elevated sensitivity and specificity. Bulbar‐onset cases had higher plasma pNfH compared to spinal onset (p = 0.0033). Furthermore, plasma pNfH positively correlated with disease progression rate (r = 0.19, p = 0.031). Baseline plasma pNfH did not influence survival in our cohort. Our findings confirmed the potential utility of plasma pNfH as a diagnostic biomarker in ALS. However, further studies with longitudinal data are needed to corroborate its prognostic value.
Collapse
Affiliation(s)
- Chiara Zecca
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy
| | - Maria Teresa Dell'Abate
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy
| | - Giuseppe Pasculli
- Department of Computer, Control, and Management Engineering Antonio Ruberti (DIAG) La Sapienza University, Rome, Italy
| | - Rosa Capozzo
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy
| | - Roberta Barone
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy
| | - Serena Arima
- Department of History, Society and Human Studies, University of Salento, Lecce, Italy
| | - Alessio Pollice
- Department of Economics and Finance, University of Bari "Aldo Moro", Lecce, Italy
| | - Vincenzo Brescia
- Unit of Laboratory Medicine, "Pia Fondazione Card.G. Panico" Hospital, Lecce, Italy
| | - Rosanna Tortelli
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy
| | - Giancarlo Logroscino
- Center for Neurodegenerative Diseases and the Aging Brain, Department of Clinical Research in Neurology, University of Bari "Aldo Moro" at "Pia Fondazione Card G. Panico" Hospital Tricase, Lecce, Italy.,Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
12
|
van der Ende EL, Bron EE, Poos JM, Jiskoot LC, Panman JL, Papma JM, Meeter LH, Dopper EGP, Wilke C, Synofzik M, Heller C, Swift IJ, Sogorb-Esteve A, Bouzigues A, Borroni B, Sanchez-Valle R, Moreno F, Graff C, Laforce R, Galimberti D, Masellis M, Tartaglia MC, Finger E, Vandenberghe R, Rowe JB, de Mendonça A, Tagliavini F, Santana I, Ducharme S, Butler CR, Gerhard A, Levin J, Danek A, Otto M, Pijnenburg YAL, Sorbi S, Zetterberg H, Niessen WJ, Rohrer JD, Klein S, van Swieten JC, Venkatraghavan V, Seelaar H. A data-driven disease progression model of fluid biomarkers in genetic frontotemporal dementia. Brain 2022; 145:1805-1817. [PMID: 34633446 PMCID: PMC9166533 DOI: 10.1093/brain/awab382] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/22/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Several CSF and blood biomarkers for genetic frontotemporal dementia have been proposed, including those reflecting neuroaxonal loss (neurofilament light chain and phosphorylated neurofilament heavy chain), synapse dysfunction [neuronal pentraxin 2 (NPTX2)], astrogliosis (glial fibrillary acidic protein) and complement activation (C1q, C3b). Determining the sequence in which biomarkers become abnormal over the course of disease could facilitate disease staging and help identify mutation carriers with prodromal or early-stage frontotemporal dementia, which is especially important as pharmaceutical trials emerge. We aimed to model the sequence of biomarker abnormalities in presymptomatic and symptomatic genetic frontotemporal dementia using cross-sectional data from the Genetic Frontotemporal dementia Initiative (GENFI), a longitudinal cohort study. Two-hundred and seventy-five presymptomatic and 127 symptomatic carriers of mutations in GRN, C9orf72 or MAPT, as well as 247 non-carriers, were selected from the GENFI cohort based on availability of one or more of the aforementioned biomarkers. Nine presymptomatic carriers developed symptoms within 18 months of sample collection ('converters'). Sequences of biomarker abnormalities were modelled for the entire group using discriminative event-based modelling (DEBM) and for each genetic subgroup using co-initialized DEBM. These models estimate probabilistic biomarker abnormalities in a data-driven way and do not rely on previous diagnostic information or biomarker cut-off points. Using cross-validation, subjects were subsequently assigned a disease stage based on their position along the disease progression timeline. CSF NPTX2 was the first biomarker to become abnormal, followed by blood and CSF neurofilament light chain, blood phosphorylated neurofilament heavy chain, blood glial fibrillary acidic protein and finally CSF C3b and C1q. Biomarker orderings did not differ significantly between genetic subgroups, but more uncertainty was noted in the C9orf72 and MAPT groups than for GRN. Estimated disease stages could distinguish symptomatic from presymptomatic carriers and non-carriers with areas under the curve of 0.84 (95% confidence interval 0.80-0.89) and 0.90 (0.86-0.94) respectively. The areas under the curve to distinguish converters from non-converting presymptomatic carriers was 0.85 (0.75-0.95). Our data-driven model of genetic frontotemporal dementia revealed that NPTX2 and neurofilament light chain are the earliest to change among the selected biomarkers. Further research should investigate their utility as candidate selection tools for pharmaceutical trials. The model's ability to accurately estimate individual disease stages could improve patient stratification and track the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Emma L van der Ende
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Esther E Bron
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jackie M Poos
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jessica L Panman
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Janne M Papma
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Elise G P Dopper
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Carolin Heller
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Imogen J Swift
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Arabella Bouzigues
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Raquel Sanchez-Valle
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, 20014 Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa, Spain
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, 17176 Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, 17176 Solna, Sweden
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, Université Laval, G1Z 1J4 Québec, Canada
| | - Daniela Galimberti
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy
- Neurodegenerative Diseases Unit, Fondazione IRCCS, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, ON M4N 3M5 Toronto, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, M5S 1A8 Toronto, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, ON N6A 3K7 London, Ontario, Canada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - James B Rowe
- Cambridge University Centre for Frontotemporal Dementia, University of Cambridge, CB2 0SZ Cambridge, UK
| | | | | | - Isabel Santana
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute and McGill University Health Centre, McGill University, 3801 Montreal, Québec, Canada
| | - Christopher R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, OX3 9DU Oxford, UK
- Department of Brain Sciences, Imperial College London, SW7 2AZ London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, M20 3LJ Manchester, UK
- Department of Nuclear Medicine and Geriatric Medicine, University Hospital Essen, 45 147 Essen, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
| | - Yolande A L Pijnenburg
- Department of Neurology, Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, 50139 Florence, Italy
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 405 30 Mölndal, Sweden
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Vikram Venkatraghavan
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
13
|
2021 White Paper on Recent Issues in Bioanalysis: ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry ( Part 2 - Recommendations on Biomarkers/CDx Assays Development & Validation, Cytometry Validation & Innovation, Biotherapeutics PK LBA Regulated Bioanalysis, Critical Reagents & Positive Controls Generation). Bioanalysis 2022; 14:627-692. [PMID: 35578974 DOI: 10.4155/bio-2022-0080] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included three Main Workshops and seven Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "context of use" [COU]); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations on ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry. Part 1A (Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC), Part 1B (Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine) and Part 3 (TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness) are published in volume 14 of Bioanalysis, issues 9 and 11 (2022), respectively.
Collapse
|
14
|
Puentes F, Benkert P, Amor S, Kuhle J, Giovannoni G. Antibodies to neurofilament light as potential biomarkers in multiple sclerosis. BMJ Neurol Open 2021; 3:e000192. [PMID: 34786556 PMCID: PMC8587694 DOI: 10.1136/bmjno-2021-000192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background and objective The concentration of neurofilament light (NfL) protein in cerebrospinal fluid (CSF) and blood is widely considered as a quantitative measure of neuro-axonal injury. Immune reactivity to NfL released into extracellular fluids induces specific autoantibody response. We investigated the levels and avidity of antibodies to NfL in patients with multiple sclerosis (MS) treated with disease-modifying therapies (DMTs) and their correlation with disease worsening and NfL protein concentration. Methods We conducted a prospective longitudinal study in 246 patients with MS (125 DMT-treated and 121 untreated at baseline). Serum levels of NfL antibodies, antibody avidity and immune complexes were determined by ELISA. NfL protein was measured using the Simoa platform. Clinical variables were tested for their association with the measured parameters in multivariate generalised estimating equation models. Results Multivariate analysis showed that levels of NfL antibodies were higher in progressive MS compared with clinically isolated syndrome (CIS)/relapsing remitting multiple sclerosis (RRMS) (p=0.010). Anti-NfL levels drop with increasing disability score (Expanded Disability Status Scale (EDSS)) (p=0.002), although conversely, were significantly elevated in CIS/RRMS after a recent EDSS increase (p=0.012). Patients receiving DMTs showed decreased levels of anti-NfL (p=0.008), high-avidity antibodies (p=0.017) and immune-complexes compared with untreated CIS/RRMS. Patients with MS switching to natalizumab showed lower levels of anti-NfL but higher immune complexes compared with healthy controls (p=0.0071). A weak association was observed between the levels of NfL protein and NfL antibodies. Conclusions These results support the potential usefulness of quantifying antibody response to NfL as potential markers of progression and treatment response in MS and need to be considered when interpreting peripheral blood NfL levels.
Collapse
Affiliation(s)
- Fabiola Puentes
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Sandra Amor
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Pathology Department, Amsterdam UMC VUMC Site, Amsterdam, The Netherlands
| | - Jens Kuhle
- Neurology, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Gavin Giovannoni
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
15
|
Dhaya I, Griton M, Konsman JP. Magnetic resonance imaging under isoflurane anesthesia alters cortical cyclooxygenase-2 expression and glial cell morphology during sepsis-associated neurological dysfunction in rats. Animal Model Exp Med 2021; 4:249-260. [PMID: 34557651 PMCID: PMC8446714 DOI: 10.1002/ame2.12167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/19/2021] [Indexed: 01/31/2023] Open
Abstract
Background Magnetic resonance imaging (MRI) of rodents combined with histology allows to determine what mechanisms underlie functional and structural brain changes during sepsis-associated encephalopathy. However, the effects of MRI performed in isoflurane-anesthetized rodents on modifications of the blood-brain barrier and the production of vasoactive prostaglandins and glia cells, which have been proposed to mediate sepsis-associated brain dysfunction, are unknown. Methods This study addressed the effect of MRI under isoflurane anesthesia on blood-brain barrier integrity, cyclooxygenase-2 expression, and glial cell activation during cecal ligature and puncture-induced sepsis-associated brain dysfunction in rats. Results Cecal ligature and puncture reduced food intake and the righting reflex. MRI under isoflurane anesthesia reduced blood-brain barrier breakdown, decreased circularity of white matter astrocytes, and increased neuronal cyclooxygenase-2 immunoreactivity in the cortex 24 hours after laparotomy. In addition, it annihilated cecal ligature and puncture-induced increased circularity of white matter microglia. MRI under isoflurane anesthesia, however, did not alter sepsis-associated perivascular cyclooxygenase-2 induction. Conclusion These findings indicate that MRI under isoflurane anesthesia of rodents can modify neurovascular and glial responses and should, therefore, be interpreted with caution.
Collapse
Affiliation(s)
- Ibtihel Dhaya
- INCIAInstitut de Neurosciences Cognitives et Intégratives d'AquitaineCNRS UMR 5287BordeauxFrance
- Univ. BordeauxINCIAUMR 5287BordeauxFrance
- Laboratoire de Neurophysiologie Fonctionnelle et PathologiesUR/11ES09Faculté des Sciences MathématiquesPhysiques et NaturellesUniversité de Tunis El ManarTunisTunisie
| | - Marion Griton
- INCIAInstitut de Neurosciences Cognitives et Intégratives d'AquitaineCNRS UMR 5287BordeauxFrance
- Univ. BordeauxINCIAUMR 5287BordeauxFrance
- Service de Réanimation Anesthésie NeurochirurgicaleCentre Hospitalier Universitaire (CHU) de BordeauxBordeauxFrance
| | - Jan Pieter Konsman
- INCIAInstitut de Neurosciences Cognitives et Intégratives d'AquitaineCNRS UMR 5287BordeauxFrance
- Univ. BordeauxINCIAUMR 5287BordeauxFrance
| |
Collapse
|
16
|
Adiutori R, Puentes F, Bremang M, Lombardi V, Zubiri I, Leoni E, Aarum J, Sheer D, McArthur S, Pike I, Malaspina A. Analysis of circulating protein aggregates as a route of investigation into neurodegenerative disorders. Brain Commun 2021; 3:fcab148. [PMID: 34396108 PMCID: PMC8361415 DOI: 10.1093/braincomms/fcab148] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/08/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022] Open
Abstract
Plasma proteome composition reflects the inflammatory and metabolic state of the organism and can be predictive of system-level and organ-specific pathologies. Circulating protein aggregates are enriched with neurofilament heavy chain-axonal proteins involved in brain aggregate formation and recently identified as biomarkers of the fatal neuromuscular disorder amyotrophic lateral sclerosis. Using unbiased proteomic methods, we have fully characterized the content in neuronal proteins of circulating protein aggregates from amyotrophic lateral sclerosis patients and healthy controls, with reference to brain protein aggregate composition. We also investigated circulating protein aggregate protein aggregation propensity, stability to proteolytic digestion and toxicity for neuronal and endothelial cell lines. Circulating protein aggregates separated by ultracentrifugation are visible as electron-dense macromolecular particles appearing as either large globular or as small filamentous formations. Analysis by mass spectrometry revealed that circulating protein aggregates obtained from patients are enriched with proteins involved in the proteasome system, possibly reflecting the underlying basis of dysregulated proteostasis seen in the disease, while those from healthy controls show enrichment of proteins involved in metabolism. Compared to the whole human proteome, proteins within circulating protein aggregates and brain aggregates show distinct chemical features of aggregation propensity, which appear dependent on the tissue or fluid of origin and not on the health status. Neurofilaments' two high-mass isoforms (460 and 268 kDa) showed a strong differential expression in amyotrophic lateral sclerosis compared to healthy control circulating protein aggregates, while aggregated neurofilament heavy chain was also partially resistant to enterokinase proteolysis in patients, demonstrated by immunoreactive bands at 171 and 31 kDa fragments not seen in digested healthy controls samples. Unbiased proteomics revealed that a total of 4973 proteins were commonly detected in circulating protein aggregates and brain, including 24 expressed from genes associated with amyotrophic lateral sclerosis. Interestingly, 285 circulating protein aggregate proteins (5.7%) were regulated (P < 0.05) and are present in biochemical pathways linked to disease pathogenesis and protein aggregation. Biologically, circulating protein aggregates from both patients and healthy controls had a more pronounced effect on the viability of hCMEC/D3 endothelial and PC12 neuronal cells compared to immunoglobulins extracted from the same plasma samples. Furthermore, circulating protein aggregates from patients exerted a more toxic effect than healthy control circulating protein aggregates on both cell lines at lower concentrations (P: 0.03, in both cases). This study demonstrates that circulating protein aggregates are significantly enriched with brain proteins which are representative of amyotrophic lateral sclerosis pathology and a potential source of biomarkers and therapeutic targets for this incurable disorder.
Collapse
Affiliation(s)
- Rocco Adiutori
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Fabiola Puentes
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Michael Bremang
- Proteome Sciences R&D GmbH & Co. KG, Frankfurt am Main 60438, Germany
| | - Vittoria Lombardi
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Irene Zubiri
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Emanuela Leoni
- Proteome Sciences R&D GmbH & Co. KG, Frankfurt am Main 60438, Germany
| | - Johan Aarum
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Denise Sheer
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Simon McArthur
- Institute of Dentistry, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Ian Pike
- Proteome Sciences plc, Hamilton House, Mabledon Place, London WC1H 9BB, UK
| | - Andrea Malaspina
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
17
|
Barro C, Zetterberg H. The blood biomarkers puzzle - A review of protein biomarkers in neurodegenerative diseases. J Neurosci Methods 2021; 361:109281. [PMID: 34237384 DOI: 10.1016/j.jneumeth.2021.109281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/07/2021] [Accepted: 07/04/2021] [Indexed: 02/04/2023]
Abstract
Neurodegenerative diseases are heterogeneous in their cause and clinical presentation making clinical assessment and disease monitoring challenging. Because of this, there is an urgent need for objective tools such as fluid biomarkers able to quantitate different aspects of the disease. In the last decade, technological improvements and awareness of the importance of biorepositories led to the discovery of an evolving number of fluid biomarkers covering the main characteristics of neurodegenerative diseases such as neurodegeneration, protein aggregates and inflammation. The ability to quantitate each aspect of the disease at a high definition enables a more precise stratification of the patients at inclusion in clinical trials, hence reducing the noise that may hamper the detection of therapeutical efficacy and allowing for smaller but likewise powered studies, which particularly improves the ability to start clinical trials for rare neurological diseases. Moreover, the use of fluid biomarkers has the potential to support a targeted therapeutical intervention, as it is now emerging for the treatment of amyloid-beta deposition in patients suffering from Alzheimer's disease. Here we review the knowledge that evolved from the measurement of fluid biomarker proteins in neurodegenerative conditions.
Collapse
Affiliation(s)
- Christian Barro
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| | - 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, Hong Kong, China
| |
Collapse
|
18
|
Spicer C, Lu CH, Catapano F, Scoto M, Zaharieva I, Malaspina A, Morgan JE, Greensmith L, Muntoni F, Zhou H. The altered expression of neurofilament in mouse models and patients with spinal muscular atrophy. Ann Clin Transl Neurol 2021; 8:866-876. [PMID: 33683023 PMCID: PMC8045929 DOI: 10.1002/acn3.51336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives To investigate the levels of neurofilaments (NFs) in transgenic mice and patients with spinal muscular atrophy (SMA), and to evaluate their efficacy as a biomarker in SMA. Methods The levels of NF mRNA transcripts were measured by quantitative real‐time PCR in spinal cord from SMA mice. Blood levels of NF heavy chain (NfH) from mice and patients were measured by an in‐house ELISA method. The response of NFs to therapeutic intervention was analysed in severe SMA mice treated with morpholino antisense oligonucleotides. Results Significant changes in NF transcript and protein in spinal cord and protein levels in blood were detected in SMA mice with severe or mild phenotypes, at different time points. A decrease in blood levels of NfH after antisense oligonucleotide treatment was only transient in the mice, despite the persistent benefit on the disease phenotype. A drastic reduction of over 90% in blood levels of NfF was observed in both control and SMA mice during early postnatal development. In contrast, blood levels of NfH were found to be decreased in older SMA children with chronic disease progression. Interpretation Our results show that blood NfH levels are informative in indicating disease onset and response to antisense oligonucleotides treatment in SMA mice, and indicate their potential as a peripheral marker reflecting the pathological status in central nervous system. In older patients with chronic SMA, however, the lower NfH levels may limit their application as biomarker, highlighting the need to continue to pursue additional biomarkers for this group of patients.
Collapse
Affiliation(s)
- Charlotte Spicer
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Ching-Hua Lu
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Neurology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Francesco Catapano
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Irina Zaharieva
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Andrea Malaspina
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jennifer E Morgan
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Linda Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - Haiyan Zhou
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom.,Genetics and Genomic Medicine Teaching and Research Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| |
Collapse
|
19
|
Zucchi E, Bonetto V, Sorarù G, Martinelli I, Parchi P, Liguori R, Mandrioli J. Neurofilaments in motor neuron disorders: towards promising diagnostic and prognostic biomarkers. Mol Neurodegener 2020; 15:58. [PMID: 33059698 PMCID: PMC7559190 DOI: 10.1186/s13024-020-00406-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Motor neuron diseases (MNDs) are etiologically and biologically heterogeneous diseases. The pathobiology of motor neuron degeneration is still largely unknown, and no effective therapy is available. Heterogeneity and lack of specific disease biomarkers have been appointed as leading reasons for past clinical trial failure, and biomarker discovery is pivotal in today's MND research agenda.In the last decade, neurofilaments (NFs) have emerged as promising biomarkers for the clinical assessment of neurodegeneration. NFs are scaffolding proteins with predominant structural functions contributing to the axonal cytoskeleton of myelinated axons. NFs are released in CSF and peripheral blood as a consequence of axonal degeneration, irrespective of the primary causal event. Due to the current availability of highly-sensitive automated technologies capable of precisely quantify proteins in biofluids in the femtomolar range, it is now possible to reliably measure NFs not only in CSF but also in blood.In this review, we will discuss how NFs are impacting research and clinical management in ALS and other MNDs. Besides contributing to the diagnosis at early stages by differentiating between MNDs with different clinical evolution and severity, NFs may provide a useful tool for the early enrolment of patients in clinical trials. Due to their stability across the disease, NFs convey prognostic information and, on a larger scale, help to stratify patients in homogenous groups. Shortcomings of NFs assessment in biofluids will also be discussed according to the available literature in the attempt to predict the most appropriate use of the biomarker in the MND clinic.
Collapse
Affiliation(s)
- Elisabetta Zucchi
- Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Bonetto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gianni Sorarù
- Neuromuscular Center, Department of Neurosciences, University of Padova, Padua, Italy.,Clinica Neurologica, Azienda Ospedaliera di Padova, Padua, Italy
| | - Ilaria Martinelli
- Department of Neurosciences, Azienda Ospedaliero Universitaria Modena, Modena, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche, Ospedale Bellaria, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche, Ospedale Bellaria, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero Universitaria Modena, Modena, Italy.
| |
Collapse
|
20
|
Lee EJ, Lim YM, Kim S, Choi L, Kim H, Kim K, Kim HW, Lee JS, Kim KK. Clinical implication of serum biomarkers and patient age in inflammatory demyelinating diseases. Ann Clin Transl Neurol 2020; 7:992-1001. [PMID: 32495489 PMCID: PMC7317646 DOI: 10.1002/acn3.51070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 04/07/2020] [Accepted: 05/03/2020] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Serum synaptic proteins levels may change with age-related neurodegeneration, affecting their clinical implications as a disease biomarker. We aimed to investigate neuronal and astroglial markers in patients with multiple sclerosis (MS) and aquaporin-4 antibody-seropositive neuromyelitis optica spectrum disorders (NMOSD) to compare the clinical implications of these markers according to age. METHODS Using single-molecule array assays, we measured neurofilament light (NfL) and glial fibrillary acidic protein (GFAP) in sera from consecutive patients with MS (n = 117) and NMOSD (n = 63). For each disease, we assessed correlations between these markers and disease severity (Expanded Disability Status Scale [EDSS]) scores according to three age groups (≤44, 45-54, and ≥55 years). RESULTS Although serum GFAP levels were significantly higher in patients with NMOSD than those with MS, levels of both serum markers revealed significant positive correlations with EDSS scores in both diseases. In MS patients, the degrees of correlation between serum NfL (or GFAP) levels and EDSS scores were similar across all age groups. However, in NMOSD patients, positive GFAP-EDSS correlations were distinctively stronger in the youngest than in the oldest group. Conversely, there were no positive NfL-EDSS correlations in NMOSD in the youngest group, but there were significant in the oldest group. INTERPRETATION The degrees to which serum NfL and GFAP levels reflect disease severity vary significantly with patient age in NMOSD, but not in MS. These findings suggest that the pathological processes and progression differ between the diseases; hence, serum biomarker levels may need to be interpreted differently according to patient age and disease type.
Collapse
Affiliation(s)
- Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Medicine, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Young-Min Lim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seungmi Kim
- Department of Medicine, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Lynkyung Choi
- Department of Medicine, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyunjin Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Keonwoo Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hye Weon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji Sung Lee
- Clinical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kwang-Kuk Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Anti-neurofilament antibodies and neurodegeneration: Markers and generators. J Neuroimmunol 2020; 344:577248. [PMID: 32344161 DOI: 10.1016/j.jneuroim.2020.577248] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 11/24/2022]
Abstract
Neuroaxonal injury and loss result in the release of cytoskeleton components, including neurofilaments, into the cerebrospinal fluid and peripheral blood. Once released, neurofilaments are highly immunogenic, inducing a specific antibody response. Anti-neurofilament antibody levels correlate with the progression of diverse neurological diseases; however, their role both in the pathogenesis of disease and as a tool for monitoring disease progression is not well understood. This study reviews the current literature on anti-neurofilament antibodies. We suggest the testing of anti-neurofilament antibodies be further developed for diagnosis and targeted for treatment.
Collapse
|
22
|
Petzold A, Lu CH, Groves M, Gobom J, Zetterberg H, Shaw G, O’Connor S. Protein aggregate formation permits millennium-old brain preservation. J R Soc Interface 2020; 17:20190775. [PMID: 31910770 PMCID: PMC7014809 DOI: 10.1098/rsif.2019.0775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/03/2019] [Indexed: 01/09/2023] Open
Abstract
Human proteins have not been reported to survive in free nature, at ambient temperature, for long periods. Particularly, the human brain rapidly dissolves after death due to auto-proteolysis and putrefaction. The here presented discovery of 2600-year-old brain proteins from a radiocarbon dated human brain provides new evidence for extraordinary long-term stability of non-amyloid protein aggregates. Immunoelectron microscopy confirmed the preservation of neurocytoarchitecture in the ancient brain, which appeared shrunken and compact compared to a modern brain. Resolution of intermediate filaments (IFs) from protein aggregates took 2-12 months. Immunoassays on micro-dissected brain tissue homogenates revealed the preservation of the known protein topography for grey and white matter for type III (glial fibrillary acidic protein, GFAP) and IV (neurofilaments, Nfs) IFs. Mass spectrometry data could be matched to a number of peptide sequences, notably for GFAP and Nfs. Preserved immunogenicity of the prehistoric human brain proteins was demonstrated by antibody generation (GFAP, Nfs, myelin basic protein). Unlike brain proteins, DNA was of poor quality preventing reliable sequencing. These long-term data from a unique ancient human brain demonstrate that aggregate formation permits for the preservation of brain proteins for millennia.
Collapse
Affiliation(s)
- Axel Petzold
- Department of Neuroinflammation and National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, UCLH, Queen Square, London WC1N 3BG, UK
- Moorfields Eye Hospital, City Road, London EC1V 2PD, UK
- Department of Neurology, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Ching-Hua Lu
- Neurology, School of Medicine, China Medical University and Hospital, Taichung City, Taiwan
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Mike Groves
- Division of Neuropathology, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg and Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg and Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Gerry Shaw
- EnCor Biotechnology Inc., 4949 SW 41st Boulevard, Ste 40., Gainesville, FL 32608, USA
| | - Sonia O’Connor
- Archaeological and Forensic Sciences, University of Bradford, Richmond Road, Bradford, West Yorkshire BD7 1DP, UK
| |
Collapse
|
23
|
Zucchi E, Lu CH, Cho Y, Chang R, Adiutori R, Zubiri I, Ceroni M, Cereda C, Pansarasa O, Greensmith L, Malaspina A, Petzold A. A motor neuron strategy to save time and energy in neurodegeneration: adaptive protein stoichiometry. J Neurochem 2019; 146:631-641. [PMID: 29959860 PMCID: PMC6175430 DOI: 10.1111/jnc.14542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 06/07/2018] [Accepted: 06/21/2018] [Indexed: 01/01/2023]
Abstract
Neurofilament proteins (Nf) are a biomarker of disease progression in amyotrophic lateral sclerosis (ALS). This study investigated whether there are major differences in expression from in vivo measurements of neurofilament isoforms, from the light chain, NfL (68 kDa), compared with larger proteins, the medium chain (NfM, 150 kDa) and the heavy (NfH, 200‐210 kDa) chains in ALS patients and healthy controls. New immunological methods were combined with Nf subunit stoichiometry calculations and Monte Carlo simulations of a coarse‐grained Nf brush model. Based on a physiological Nf subunit stoichiometry of 7 : 3 : 2 (NfL:NfM:NfH), we found an ‘adaptive’ Nf subunit stoichiometry of 24 : 2.4 : 1.6 in ALS. Adaptive Nf stoichiometry preserved NfL gyration radius in the Nf brush model. The energy and time requirements for Nf translation were 56 ± 27k ATP (5.6 h) in control subjects compared to 123 ± 102k (12.3 h) in ALS with ‘adaptive’ (24:2.4:1.6) Nf stoichiometry (not significant) and increased significantly to 355 ± 330k (35.5 h) with ‘luxury’ (7:3:2) Nf subunit stoichiometry (p < 0.0001 for each comparison). Longitudinal disease progression‐related energy consumption was highest with a ‘luxury’ (7:3:2) Nf stoichiometry. Therefore, an energy and time‐saving option for motor neurons is to shift protein expression from larger to smaller (cheaper) subunits, at little or no costs on a protein structural level, to compensate for increased energy demands. ![]()
Collapse
Affiliation(s)
- Elisabetta Zucchi
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Center of Genomic and post-Genomic, IRCCS Mondino Foundation, Pavia, Italy
| | - Ching-Hua Lu
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Neurology, China Medical University Hospital, Taichung City, Taiwan
| | - Yunju Cho
- Department of Chemistry, Kwangwoon University, Seoul, Korea
| | - Rakwoo Chang
- Department of Chemistry, Kwangwoon University, Seoul, Korea
| | - Rocco Adiutori
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Irene Zubiri
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Ceroni
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,General Neurology Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Cereda
- Center of Genomic and post-Genomic, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Center of Genomic and post-Genomic, IRCCS Mondino Foundation, Pavia, Italy
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, University College London, London, UK
| | - Andrea Malaspina
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Axel Petzold
- Department of Neuromuscular Diseases, MRC Centre for Neuromuscular Diseases, Queen Square, London, UK.,The National Hospital for Neurology and Neurosurgery, Queen Square, London, UK.,Moorfields Eye Hospital, London, UK.,Amsterdam UMC, Departments of Neurology and Ophthalmology, De Boelelaan, Amsterdam, NL
| |
Collapse
|
24
|
Kušnierová P, Zeman D, Hradílek P, Čábal M, Zapletalová O. Neurofilament levels in patients with neurological diseases: A comparison of neurofilament light and heavy chain levels. J Clin Lab Anal 2019; 33:e22948. [PMID: 31199010 PMCID: PMC6757126 DOI: 10.1002/jcla.22948] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/26/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
Background Neurofilaments are the major cytoskeletal components of neurons, and cell injury leads to their release into the surrounding area. The aim of this study was to compare the cerebrospinal fluid (CSF) and serum (S) concentrations of neurofilament light chains (NFLs) and phosphorylated neurofilament heavy chains (pNFHs). Methods Neurofilament concentrations were measured in CSF and S samples from 172 patients using three enzyme‐linked immunosorbent assays. Excel, Stata version 13, MedCal version 17.9.7., and NCSS 2007 software were used for the statistical analysis. Results There was a statistically significant correlation between the concentrations of CSF NFL and CSF pNFH (rs = 0.748; n = 89; P < 0.001), but Passing‐Bablok regression showed systematic deviation between the values obtained using the two assays. This indicates that the assays were not interchangeable. CSF pNFH and S pNFH concentrations showed low correlation. The kappa statistic showed moderate conformity between CSF pNFH and CSF NFL concentrations (κ = 0.556). Conclusions The CSF NFL and CSF pNFH assays gave clinically consistent results that reflected the degree of axonal damage, independent of any particular neurological diagnosis. The S pNFH assays had a lower predictive value due to the low correlation coefficient and the kappa index of the CSF pNFH method.
Collapse
Affiliation(s)
- Pavlína Kušnierová
- Department of Clinical Biochemistry, Institute of Laboratory Diagnostics, University Hospital Ostrava, Ostrava, Czech Republic
| | - David Zeman
- Department of Clinical Biochemistry, Institute of Laboratory Diagnostics, University Hospital Ostrava, Ostrava, Czech Republic.,Clinic of Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Pavel Hradílek
- Clinic of Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Martin Čábal
- Clinic of Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Olga Zapletalová
- Clinic of Neurology, University Hospital Ostrava, Ostrava, Czech Republic
| |
Collapse
|
25
|
Adiutori R, Aarum J, Zubiri I, Bremang M, Jung S, Sheer D, Pike I, Malaspina A. The proteome of neurofilament-containing protein aggregates in blood. Biochem Biophys Rep 2018; 14:168-177. [PMID: 29872749 PMCID: PMC5986704 DOI: 10.1016/j.bbrep.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/15/2018] [Accepted: 04/26/2018] [Indexed: 11/24/2022] Open
Abstract
Protein aggregation in biofluids is a poorly understood phenomenon. Under normal physiological conditions, fluid-borne aggregates may contain plasma or cell proteins prone to aggregation. Recent observations suggest that neurofilaments (Nf), the building blocks of neurons and a biomarker of neurodegeneration, are included in high molecular weight complexes in circulation. The composition of these Nf-containing hetero-aggregates (NCH) may change in systemic or organ-specific pathologies, providing the basis to develop novel disease biomarkers. We have tested ultracentrifugation (UC) and a commercially available protein aggregate binder, Seprion PAD-Beads (SEP), for the enrichment of NCH from plasma of healthy individuals, and then characterised the Nf content of the aggregate fractions using gel electrophoresis and their proteome by mass spectrometry (MS). Western blot analysis of fractions obtained by UC showed that among Nf isoforms, neurofilament heavy chain (NfH) was found within SDS-stable high molecular weight aggregates. Shotgun proteomics of aggregates obtained with both extraction techniques identified mostly cell structural and to a lesser extent extra-cellular matrix proteins, while functional analysis revealed pathways involved in inflammatory response, phagosome and prion-like protein behaviour. UC aggregates were specifically enriched with proteins involved in endocrine, metabolic and cell-signalling regulation. We describe the proteome of neurofilament-containing aggregates isolated from healthy individuals biofluids using different extraction methods.
Collapse
Affiliation(s)
- Rocco Adiutori
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| | - Johan Aarum
- Centre for Genomics and Child Health, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Irene Zubiri
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| | - Michael Bremang
- Proteome Sciences Plc, Hamilton House, Mabledon Place, London, United Kingdom
| | - Stephan Jung
- ProteomeSciencesR&DGmbH&Co.KG, Frankfurt, Germany
| | - Denise Sheer
- Centre for Genomics and Child Health, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Ian Pike
- Proteome Sciences Plc, Hamilton House, Mabledon Place, London, United Kingdom
| | - Andrea Malaspina
- Centre for Neuroscience and Trauma, Queen Mary University of London, Blizard Institute, Barts and The School of Medicine and Dentistry, London, United Kingdom
| |
Collapse
|
26
|
Gong ZY, Lv GP, Gao LN, Lu Y, Guo J, Zang DW. Neurofilament Subunit L Levels in the Cerebrospinal Fluid and Serum of Patients with Amyotrophic Lateral Sclerosis. NEURODEGENER DIS 2018; 18:165-172. [PMID: 29898446 DOI: 10.1159/000488681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 03/20/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND There are no reliable biomarkers that could evaluate the disease burden in amyotrophic lateral sclerosis (ALS). OBJECTIVES The aim of our study is to evaluate the changes in cerebrospinal fluid (CSF) and serum neurofilament subunit L (NF-L) in patients with ALS and to analyze the correlations between the levels of NF-L and clinical parameters. METHOD CSF and serum samples were obtained from 80 ALS patients and 40 controls. The levels of NF-L in CSF and serum were assessed, and disease progression parameters including duration, revised ALS Functional Rating Scale (ALSFRS-r) score, disease progression rate (DPR), upper motor neuron (UMN) score, and survival were analyzed by registered neurologists. All samples were measured using a commercial enzyme-linked immunosorbent assay. Statistical analyses were performed using Prism software. RESULTS Compared to the controls, the ALS patients displayed significantly increased levels of NF-L; these values were negatively correlated with the ALSFRS-r score and positively correlated with the decrease in ALSFRS-r score, DPR, and UMN score. There was no correlation between levels of NF-L and duration. In addition, the cumulative survival rate in ALS patients with a low level of NF-L was higher than in patients with a high level of NF-L. CONCLUSIONS NF-L levels increased in CSF and serum of patients with ALS. NF-L may thus be a neurodegenerative biomarker for predicting ALS severity and progression, and the survival of patients with this disease.
Collapse
Affiliation(s)
- Zhong-Ying Gong
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| | - Gao-Peng Lv
- Department of Neurology, Tianjin First Center Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li-Na Gao
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| | - Yi Lu
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| | - Jie Guo
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| | - Da-Wei Zang
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
De Schaepdryver M, Jeromin A, Gille B, Claeys KG, Herbst V, Brix B, Van Damme P, Poesen K. Comparison of elevated phosphorylated neurofilament heavy chains in serum and cerebrospinal fluid of patients with amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2018; 89:367-373. [PMID: 29054919 DOI: 10.1136/jnnp-2017-316605] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Phosphorylated neurofilament heavy chain (pNfH) levels are elevated in cerebrospinal fluid (CSF) of patients with amyotrophic lateral sclerosis (ALS). Instead of CSF, we explored blood as an alternative source to measure pNfH in patients with ALS. METHODS In this single centre retrospective study, 85 patients with ALS, 215 disease controls (DC) and 31 ALS mimics were included. Individual serum pNfH concentrations were correlated with concentrations in CSF and with several clinical parameters. The performance characteristics of pNfH in CSF and serum of patients with ALS and controls were calculated and compared using receiver operating characteristic (ROC) curves. RESULTS CSF and serum pNfH concentrations in patients with ALS correlated well (r=0.652, p<0.0001) and were significantly increased compared with DC (p<0.0001) and ALS mimics (p<0.0001). CSF pNfH outperformed serum pNfH in discriminating patients with ALS from DC and ALS mimics (difference between area under the ROC curves: p=0.0001 and p=0.0005; respectively). Serum pNfH correlated inversely with symptom duration (r=-0.315, p=0.0033). CSF and serum pNfH were lower when the disease progression rate was slower (r=0.279, p<0.01 and r=0.289, p<0.01; respectively). Unlike CSF, serum pNfH did not correlate with the burden of clinical and electromyographic motor neuron dysfunction. CONCLUSIONS CSF and serum pNfH concentrations are elevated in patients with ALS and correlate with the disease progression rate. Moreover, CSF pNfH correlates with the burden of motor neuron dysfunction. Our findings encourage further pursuit of CSF and serum pNfH concentrations in the diagnostic pathway of patients suspected to have ALS.
Collapse
Affiliation(s)
- Maxim De Schaepdryver
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, KU Leuven (University of Leuven), Leuven, Belgium.,Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | - Benjamin Gille
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Department of Neurosciences, Laboratory for Muscle diseases and Neuropathies, KU Leuven (University of Leuven), Leuven, Belgium
| | | | | | - Philip Van Damme
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Department of Neurosciences, KU Leuven (University of Leuven) and Center for Brain & Disease Research VIB Leuven, Leuven, Belgium
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, KU Leuven (University of Leuven), Leuven, Belgium.,Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Sandelius Å, Zetterberg H, Blennow K, Adiutori R, Malaspina A, Laura M, Reilly MM, Rossor AM. Plasma neurofilament light chain concentration in the inherited peripheral neuropathies. Neurology 2018; 90:e518-e524. [PMID: 29321234 PMCID: PMC5818017 DOI: 10.1212/wnl.0000000000004932] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/17/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To perform a cross-sectional study to determine whether plasma neurofilament light chain (NfL) concentration is elevated in patients with Charcot-Marie-Tooth disease (CMT) and if it correlates with disease severity. METHODS Blood samples were collected from 75 patients with CMT and 67 age-matched healthy controls over a 1-year period. Disease severity was measured using the Rasch modified CMT Examination and neuropathy scores. Plasma NfL concentration was measured using an in-house-developed Simoa assay. RESULTS Plasma NfL concentration was significantly higher in patients with CMT (median 26.0 pg/mL) compared to healthy controls (median 14.6 pg/mL, p < 0.0001) and correlated with disease severity as measured using the Rasch modified CMT examination (r = 0.43, p < 0.0001) and neuropathy (r = 0.37, p = 0.044) scores. Concentrations were also significantly higher when subdividing patients by genetic subtype (CMT1A, SPTLC1, and GJB1) or into demyelinating or axonal forms compared to healthy controls. CONCLUSION There are currently no validated blood biomarkers for peripheral neuropathy. The significantly raised plasma NfL concentration in patients with CMT and its correlation with disease severity suggest that plasma NfL holds promise as a biomarker of disease activity, not only for inherited neuropathies but for peripheral neuropathy in general.
Collapse
Affiliation(s)
- Åsa Sandelius
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Henrik Zetterberg
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Kaj Blennow
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Rocco Adiutori
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Andrea Malaspina
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Matilde Laura
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Mary M Reilly
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK.
| | - Alexander M Rossor
- From the Department of Psychiatry and Neurochemistry (Å.S., H.Z., K.B.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology; Trauma and Neuroscience Centre (R.A., A.M.), Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London; and MRC Centre for Neuromuscular Diseases (M.L., M.M.R., A.M.R.), UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
29
|
|
30
|
Neurofilaments as Biomarkers for Amyotrophic Lateral Sclerosis: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0164625. [PMID: 27732645 PMCID: PMC5061412 DOI: 10.1371/journal.pone.0164625] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To allow early diagnosis and monitoring of disease progression, there is a need for biomarkers in amyotrophic lateral sclerosis (ALS). Neurofilaments (NF) are emerging protein biomarkers in other neurological diseases, and are of possible use in ALS. OBJECTIVE The aim of this study is to evaluate the utility of NF levels as blood or cerebrospinal fluid (CSF) biomarker in patients with ALS. METHODS A systematic search of Pubmed, Embase and Scopus was performed. Methodological quality assessment was applied to refine the final search results. Meta-analysis of the data was performed. RESULTS Level of NF heavy chain and light chains were significantly elevated in the CSF of ALS patients compared to healthy controls/controls without parenchymal central nervous system (CNS) involvement and ALS mimic disease patients. NF light chain level in CSF was higher in ALS patients than in neurological patients with CNS involvement (SMD = 1.352, P = 0.01). NF light chain concentration in blood was higher in ALS patients than healthy controls/controls without CNS involvement (SMD = 1.448, P<0.0001). NF heavy chain levels in CSF were negatively correlated disease duration and ALSFRS-R ((r = -0.447, P<0.0001; r = -0.486, P<0.0001). NF light chain levels in CSF were negatively correlated with disease duration (r = -0.273, P = 0.011). CONCLUSION NF heavy and light chain levels have potential use as a marker of neural degeneration in ALS, but are not specific for the disease, and are more likely to be used as measures of disease progression.
Collapse
|
31
|
Poulsen NN, Pedersen ME, Østergaard J, Petersen NJ, Nielsen CT, Heegaard NHH, Jensen H. Flow-Induced Dispersion Analysis for Probing Anti-dsDNA Antibody Binding Heterogeneity in Systemic Lupus Erythematosus Patients: Toward a New Approach for Diagnosis and Patient Stratification. Anal Chem 2016; 88:9056-61. [DOI: 10.1021/acs.analchem.6b01741] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Nicklas N. Poulsen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Morten E. Pedersen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Jesper Østergaard
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Nickolaj J. Petersen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Christoffer T. Nielsen
- Department of Autoimmunology & Biomarkers, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Niels H. H. Heegaard
- Department of Autoimmunology & Biomarkers, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
- Department
of Clinicial Biochemistry, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Henrik Jensen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
32
|
Li S, Ren Y, Zhu W, Yang F, Zhang X, Huang X. Phosphorylated neurofilament heavy chain levels in paired plasma and CSF of amyotrophic lateral sclerosis. J Neurol Sci 2016; 367:269-74. [DOI: 10.1016/j.jns.2016.05.062] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/10/2016] [Accepted: 05/31/2016] [Indexed: 12/01/2022]
|
33
|
Rossor AM, Lu CH, Petzold A, Malaspina A, Laura M, Greensmith L, Reilly MM. Plasma neurofilament heavy chain is not a useful biomarker in Charcot-Marie-Tooth disease. Muscle Nerve 2016; 53:972-5. [PMID: 27015106 DOI: 10.1002/mus.25124] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The negative results in trials of vitamin C in Charcot-Marie-Tooth disease (CMT) type 1A have highlighted the lack of sensitive outcome measures. Neurofilaments are abundant neuronal cytoskeletal proteins, and their concentration in blood is likely to reflect axonal breakdown. We therefore examined plasma neurofilament heavy-chain (NfH) concentration as a potential biomarker in CMT. METHODS Blood samples were collected from healthy controls and patients with CMT over a 2-year period. Disease severity was measured using the CMT Examination Score. An in-house enzyme-linked immunoabsorbent assay was used to measure plasma NfH levels. RESULTS There was no significant difference in plasma NfH concentrations between CMT patients and controls (P = 0.449). There was also no significant difference in plasma NfH levels in the CMT group over 1 year (mean difference = -0.02, SEM = 4.44, P = 0.98). CONCLUSIONS Plasma NfH levels are not altered in patients with CMT and are not a suitable biomarker of disease activity. Muscle Nerve 53: 972-975, 2016.
Collapse
Affiliation(s)
- Alexander M Rossor
- Medical Research Council Centre for Neuromuscular Diseases, University College London Institute of Neurology and National Hospital for Neurology and Neurosurgery, 811 Queen Square, London, WC1N 3BG, UK
| | - Ching-Hua Lu
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London Institute of Neurology, London, UK.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Axel Petzold
- Department of Neurology, VU Medical Centre, Amsterdam, The Netherlands
| | - Andreas Malaspina
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matilde Laura
- Medical Research Council Centre for Neuromuscular Diseases, University College London Institute of Neurology and National Hospital for Neurology and Neurosurgery, 811 Queen Square, London, WC1N 3BG, UK
| | - Linda Greensmith
- Medical Research Council Centre for Neuromuscular Diseases, University College London Institute of Neurology and National Hospital for Neurology and Neurosurgery, 811 Queen Square, London, WC1N 3BG, UK
| | - Mary M Reilly
- Medical Research Council Centre for Neuromuscular Diseases, University College London Institute of Neurology and National Hospital for Neurology and Neurosurgery, 811 Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
34
|
CSF and Blood Levels of GFAP in Alexander Disease. eNeuro 2015; 2:eN-NWR-0080-15. [PMID: 26478912 PMCID: PMC4603256 DOI: 10.1523/eneuro.0080-15.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 01/28/2023] Open
Abstract
Alexander disease is a rare, progressive, and generally fatal neurological disorder that results from dominant mutations affecting the coding region of GFAP, the gene encoding glial fibrillary acidic protein, the major intermediate filament protein of astrocytes in the CNS. A key step in pathogenesis appears to be the accumulation of GFAP within astrocytes to excessive levels. Studies using mouse models indicate that the severity of the phenotype correlates with the level of expression, and suppression of GFAP expression and/or accumulation is one strategy that is being pursued as a potential treatment. With the goal of identifying biomarkers that indirectly reflect the levels of GFAP in brain parenchyma, we have assayed GFAP levels in two body fluids in humans that are readily accessible as biopsy sites: CSF and blood. We find that GFAP levels are consistently elevated in the CSF of patients with Alexander disease, but only occasionally and modestly elevated in blood. These results provide the foundation for future studies that will explore whether GFAP levels can serve as a convenient means to monitor the progression of disease and the response to treatment.
Collapse
|
35
|
Lu CH, Macdonald-Wallis C, Gray E, Pearce N, Petzold A, Norgren N, Giovannoni G, Fratta P, Sidle K, Fish M, Orrell R, Howard R, Talbot K, Greensmith L, Kuhle J, Turner MR, Malaspina A. Neurofilament light chain: A prognostic biomarker in amyotrophic lateral sclerosis. Neurology 2015; 84:2247-57. [PMID: 25934855 PMCID: PMC4456658 DOI: 10.1212/wnl.0000000000001642] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/20/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test blood and CSF neurofilament light chain (NfL) levels in relation to disease progression and survival in amyotrophic lateral sclerosis (ALS). METHODS Using an electrochemiluminescence immunoassay, NfL levels were measured in samples from 2 cohorts of patients with sporadic ALS and healthy controls, recruited in London (ALS/control, plasma: n = 103/42) and Oxford (ALS/control, serum: n = 64/36; paired CSF: n = 38/20). NfL levels in patients were measured at regular intervals for up to 3 years. Change in ALS Functional Rating Scale-Revised score was used to assess disease progression. Survival was evaluated using Cox regression and Kaplan-Meier analysis. RESULTS CSF, serum, and plasma NfL discriminated patients with ALS from healthy controls with high sensitivity (97%, 89%, 90%, respectively) and specificity (95%, 75%, 71%, respectively). CSF NfL was highly correlated with serum levels (r = 0.78, p < 0.0001). Blood NfL levels were approximately 4 times as high in patients with ALS compared with controls in both cohorts, and maintained a relatively constant expression during follow-up. Blood NfL levels at recruitment were strong, independent predictors of survival. The highest tertile of blood NfL at baseline had a mortality hazard ratio of 3.91 (95% confidence interval 1.98-7.94, p < 0.001). CONCLUSION Blood-derived NfL level is an easily accessible biomarker with prognostic value in ALS. The individually relatively stable levels longitudinally offer potential for NfL as a pharmacodynamic biomarker in future therapeutic trials. CLASSIFICATION OF EVIDENCE This report provides Class III evidence that the NfL electrochemiluminescence immunoassay accurately distinguishes patients with sporadic ALS from healthy controls.
Collapse
Affiliation(s)
- Ching-Hua Lu
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Corrie Macdonald-Wallis
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Elizabeth Gray
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Neil Pearce
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Axel Petzold
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Niklas Norgren
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Gavin Giovannoni
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Pietro Fratta
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Katie Sidle
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Mark Fish
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Richard Orrell
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Robin Howard
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Kevin Talbot
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Linda Greensmith
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Jens Kuhle
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK
| | - Martin R Turner
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK.
| | - Andrea Malaspina
- From the Centre for Neuroscience & Trauma (C.-H.L., G.G., J.K., A.M.), Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Sobell Department of Motor Neuroscience and Movement Disorders (C.-H.L., L.G.), Departments of Neuroinflammation (A.P.), Neurodegenerative Disease (P.F.), Molecular Neuroscience (K.S.), and Clinical Neuroscience (R.O.), and MRC Centre for Neuromuscular Diseases (R.O., L.G.), UCL Institute of Neurology, London; MRC Integrative Epidemiology Unit (C.M.-W.), University of Bristol; Nuffield Department of Clinical Neurosciences (E.G., K.T., M.R.T.), University of Oxford; Department of Medical Statistics (N.P.), London School of Hygiene and Tropical Medicine, London, UK; UmanDiagnostics (N.N.), Umeå, Sweden; Medicine Clinical Trial Unit (M.F.), Musgrove Park Hospital, Taunton, UK; National Hospital for Neurology and Neurosurgery (R.O., R.H., A.M.), London, UK; Neurology (J.K.), University Hospital Basel, Switzerland; North-East London and Essex MND Care and Research Centre (A.M.), London; and Basildon and Thurrock University Hospitals NHS Foundation Trust (A.M.), Basildon, UK.
| |
Collapse
|
36
|
Lu CH, Petzold A, Topping J, Allen K, Macdonald-Wallis C, Clarke J, Pearce N, Kuhle J, Giovannoni G, Fratta P, Sidle K, Fish M, Orrell R, Howard R, Greensmith L, Malaspina A. Plasma neurofilament heavy chain levels and disease progression in amyotrophic lateral sclerosis: insights from a longitudinal study. J Neurol Neurosurg Psychiatry 2015; 86:565-73. [PMID: 25009280 PMCID: PMC4413806 DOI: 10.1136/jnnp-2014-307672] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/16/2014] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To investigate the role of longitudinal plasma neurofilament heavy chain protein (NfH) levels as an indicator of clinical progression and survival in amyotrophic lateral sclerosis (ALS). METHODS A cross-sectional study involving 136 clinically heterogeneous patients with ALS and 104 healthy and neurological controls was extended to include a prospective analysis of 74 of these ALS cases, with samplings at approximately 3-month intervals in a follow-up period of up to 3 years. We analysed the correlation between longitudinal NfH-phosphoform levels and disease progression. Temporal patterns of NfH changes were evaluated using multilevel linear regression. RESULTS Baseline plasma NfH levels were higher than controls only in patients with ALS with short disease duration to baseline sampling. Compared with controls, fast-progressing patients with ALS, particularly those with a short diagnostic latency and disease duration, had higher plasma NfH levels at an early stage and lower levels closer to end-stage disease. Lower NfH levels between visits were associated with rapid functional deterioration. We also detected antibodies against NfH, NfH aggregates and NfH cleavage products. CONCLUSIONS Disease progression in ALS involves defined trajectories of plasma NfH levels, reflecting speed of neurological decline and survival. Intervisit plasma NfH changes are also indicative of disease progression. This study confirms that longitudinal measurements of NfH plasma levels are more informative than cross-sectional studies, where the time of sampling may represent a bias in the interpretation of the results. Autoantibodies against NfH aggregates and NfH cleavage products may explain the variable expression of plasma NfH with disease progression. TRAIL REGISTRATION NUMBER NIHRID6160.
Collapse
Affiliation(s)
- Ching-Hua Lu
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK
| | - Axel Petzold
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | - Jo Topping
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kezia Allen
- Basildon and Thurrock University Hospitals NHS Foundation Trust, Essex, UK
| | | | - Jan Clarke
- National Hospital for Neurology and Neurosurgery, London, UK
| | - Neil Pearce
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Jens Kuhle
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gavin Giovannoni
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Pietro Fratta
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Katie Sidle
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Mark Fish
- Clinical Trial Unit, Musgrove Park Hospital, Taunton, UK
| | - Richard Orrell
- National Hospital for Neurology and Neurosurgery, London, UK Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK
| | - Robin Howard
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - Andrea Malaspina
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK Basildon and Thurrock University Hospitals NHS Foundation Trust, Essex, UK North-East London and Essex MND Care and Research Centre, London, UK
| |
Collapse
|
37
|
McCombe PA, Pfluger C, Singh P, Lim CYH, Airey C, Henderson RD. Serial measurements of phosphorylated neurofilament-heavy in the serum of subjects with amyotrophic lateral sclerosis. J Neurol Sci 2015; 353:122-9. [PMID: 25958264 DOI: 10.1016/j.jns.2015.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/31/2022]
Abstract
There is a need for a blood biomarker of disease activity in ALS. This marker needs to measure the loss of motor neurones. Phosphorylated neurofilament heavy chain (pNfH) in the serum is a biomarker of axonal injury. Previous studies have found that levels of pNfH are elevated in ALS. We have performed a serial study of pNfH levels in 98 subjects from our ALS clinic. There was significant elevation of levels of pNfH in subjects with ALS compared to controls, although there was considerable variability. In studies of individuals who had two or more serial samples, we found that the levels of pNfH increased over time in the early stage of disease. Levels were low in subjects with long survival. The rate of rise of pNfH was inversely correlated with survival. We suggest that the initial level of pNfH is a marker of disease severity and that changes in pNfH levels are markers of disease progression.
Collapse
Affiliation(s)
- P A McCombe
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia.
| | - C Pfluger
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - P Singh
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - C Y H Lim
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - C Airey
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| | - R D Henderson
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane Hospital, Herston, Queensland 4029, Australia
| |
Collapse
|
38
|
Logroscino G, Tortelli R, Rizzo G, Marin B, Preux PM, Malaspina A. Amyotrophic Lateral Sclerosis: An Aging-Related Disease. CURRENT GERIATRICS REPORTS 2015. [DOI: 10.1007/s13670-015-0127-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Petzold A. Glial fibrillary acidic protein is a body fluid biomarker for glial pathology in human disease. Brain Res 2015; 1600:17-31. [DOI: 10.1016/j.brainres.2014.12.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/01/2014] [Indexed: 12/20/2022]
|
40
|
Puentes F, Topping J, Kuhle J, van der Star BJ, Douiri A, Giovannoni G, Baker D, Amor S, Malaspina A. Immune reactivity to neurofilament proteins in the clinical staging of amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2014; 85:274-8. [PMID: 24078718 DOI: 10.1136/jnnp-2013-305494] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Neurofilament (NF) proteins detection in biological fluids as a by-product of axonal loss is technically challenging and to date relies mostly on cerebrospinal fluid (CSF) measurements. Plasma antibodies against NF proteins and particularly to their soluble light chain (NF-L) could be a more practical surrogate marker for disease staging in amyotrophic lateral sclerosis (ALS), an invariably fatal and clinically heterogeneous neuromuscular disorder. METHODOLOGY We have used a recombinant neurofilament light chain (NF-L) protein for the ELISA detection of antibodies against NF proteins in plasma samples from a well-characterised cohort of ALS individuals (n:73). The use of an established functional rating scale and of a recently proposed staging of disease progression allowed stratification of the ALS cohort based on disease stage, site of onset, survival and speed of disease progression. RESULTS Antibody levels to NF proteins in plasma were significantly higher in ALS individuals compared to healthy controls (p<0.001). Higher NF plasma immunoreactivity was seen in advanced ALS cases (stage IVA-B) compared to earlier phases of the disease (p<0.05). There was no difference in anti-NF plasma antibodies between ALS individuals treated with riluzole and untreated patients; although riluzole-treated ALS cases with an earlier age of onset and with a shorter diagnostic delay displayed higher anti-NFL antibody levels compared to untreated ALS patients with similar features. CONCLUSIONS Immunoreactivity to plasma NF-L and homologous NF proteins is informative of the stage of disease progression in ALS. The determination of NF antibody levels in plasma could be added to the growing panel of disease-monitoring biomarkers in ALS targeting cytoskeletal antigens.
Collapse
Affiliation(s)
- Fabiola Puentes
- Neuroimmunology Unit, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, , London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Neurofilament light antibodies in serum reflect response to natalizumab treatment in multiple sclerosis. Mult Scler 2014; 20:1355-62. [DOI: 10.1177/1352458514521887] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background: Increased levels of antibodies to neurofilament light protein (NF-L) in biological fluids have been found to reflect neuroinflammatory responses and neurodegeneration in multiple sclerosis (MS). Objective: To evaluate whether levels of serum antibodies against NF-L correlate with clinical variants and treatment response in MS. Methods: The autoantibody reactivity to NF-L protein was tested in serum samples from patients with relapsing–remitting MS (RRMS) ( n=22) and secondary progressive MS (SPMS) ( n=26). Two other cohorts of RRMS patients under treatment with natalizumab were analysed cross-sectionally ( n=16) and longitudinally ( n=24). The follow-up samples were taken at 6, 12, 18 and 24 months after treatment, and the NF-L antibody levels were compared against baseline levels. Results: NF-L antibodies were higher in MS clinical groups than healthy controls and in RRMS compared to SPMS patients ( p<0.001). NF-L antibody levels were lower in natalizumab treated than in untreated patients ( p<0.001). In the longitudinal series, NF-L antibody levels decreased over time and a significant difference was found following 24 months of treatment compared with baseline measurements ( p=0.001). Conclusions: Drug efficacy in MS treatment indicates the potential use of monitoring the content of antibodies against the NF-L chain as a predictive biomarker of treatment response in MS.
Collapse
|
42
|
Stevenson L, Kelley M, Gorovits B, Kingsley C, Myler H, Osterlund K, Muruganandam A, Minamide Y, Dominguez M. Large molecule specific assay operation: recommendation for best practices and harmonization from the global bioanalysis consortium harmonization team. AAPS J 2014; 16:83-8. [PMID: 24242296 PMCID: PMC3889533 DOI: 10.1208/s12248-013-9542-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 10/15/2013] [Indexed: 11/30/2022] Open
Abstract
The L2 Global Harmonization Team on large molecule specific assay operation for protein bioanalysis in support of pharmacokinetics focused on the following topics: setting up a balanced validation design, specificity testing, selectivity testing, dilutional linearity, hook effect, parallelism, and testing of robustness and ruggedness. The team additionally considered the impact of lipemia, hemolysis, and the presence of endogenous analyte on selectivity assessments as well as the occurrence of hook effect in study samples when no hook effect had been observed during pre-study validation.
Collapse
Affiliation(s)
- Lauren Stevenson
- Biogen Idec, 14 Cambridge Center, Cambridge, Massachusetts, 02142, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Gaiottino J, Norgren N, Dobson R, Topping J, Nissim A, Malaspina A, Bestwick JP, Monsch AU, Regeniter A, Lindberg RL, Kappos L, Leppert D, Petzold A, Giovannoni G, Kuhle J. Increased neurofilament light chain blood levels in neurodegenerative neurological diseases. PLoS One 2013; 8:e75091. [PMID: 24073237 PMCID: PMC3779219 DOI: 10.1371/journal.pone.0075091] [Citation(s) in RCA: 354] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/08/2013] [Indexed: 11/25/2022] Open
Abstract
Objective Neuronal damage is the morphological substrate of persisting neurological disability. Neurofilaments (Nf) are cytoskeletal proteins of neurons and their release into cerebrospinal fluid has shown encouraging results as a biomarker for neurodegeneration. This study aimed to validate the quantification of the Nf light chain (NfL) in blood samples, as a biofluid source easily accessible for longitudinal studies. Methods We developed and applied a highly sensitive electrochemiluminescence (ECL) based immunoassay for quantification of NfL in blood and CSF. Results Patients with Alzheimer’s disease (AD) (30.8 pg/ml, n=20), Guillain-Barré-syndrome (GBS) (79.4 pg/ml, n=19) or amyotrophic lateral sclerosis (ALS) (95.4 pg/ml, n=46) had higher serum NfL values than a control group of neurological patients without evidence of structural CNS damage (control patients, CP) (4.4 pg/ml, n=68, p<0.0001 for each comparison, p=0.002 for AD patients) and healthy controls (HC) (3.3 pg/ml, n=67, p<0.0001). Similar differences were seen in corresponding CSF samples. CSF and serum levels correlated in AD (r=0.48, p=0.033), GBS (r=0.79, p<0.0001) and ALS (r=0.70, p<0.0001), but not in CP (r=0.11, p=0.3739). The sensitivity and specificity of serum NfL for separating ALS from healthy controls was 91.3% and 91.0%. Conclusions We developed and validated a novel ECL based sandwich immunoassay for the NfL protein in serum (NfLUmea47:3); levels in ALS were more than 20-fold higher than in controls. Our data supports further longitudinal studies of serum NfL in neurodegenerative diseases as a potential biomarker of on-going disease progression, and as a potential surrogate to quantify effects of neuroprotective drugs in clinical trials.
Collapse
Affiliation(s)
- Johanna Gaiottino
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Bone & Joint Research Unit, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | | | - Ruth Dobson
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Joanne Topping
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ahuva Nissim
- Bone & Joint Research Unit, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Andrea Malaspina
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- North-East London and Essex Regional MND Care Centre, London, United Kingdom
| | - Jonathan P. Bestwick
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andreas U. Monsch
- Memory Clinic, Department of Geriatrics, University Hospital, Basel, Basel, Switzerland
| | - Axel Regeniter
- Laboratory Medicine, University Hospital, Basel, Basel, Switzerland
| | - Raija L. Lindberg
- Department of Neurology, University Hospital, Basel, Basel, Switzerland
| | - Ludwig Kappos
- Department of Neurology, University Hospital, Basel, Basel, Switzerland
| | - David Leppert
- Department of Neurology, University Hospital, Basel, Basel, Switzerland
| | - Axel Petzold
- UCL Institute of Neurology, Department of Neuroinflammation, London, United Kingdom
| | - Gavin Giovannoni
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jens Kuhle
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Bone & Joint Research Unit, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
- Department of Neurology, University Hospital, Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
45
|
Kuhle J, Plattner K, Bestwick JP, Lindberg RL, Ramagopalan SV, Norgren N, Nissim A, Malaspina A, Leppert D, Giovannoni G, Kappos L. A comparative study of CSF neurofilament light and heavy chain protein in MS. Mult Scler 2013; 19:1597-603. [PMID: 23529999 DOI: 10.1177/1352458513482374] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND There is a lack of reliable biomarkers of axonal degeneration. Neurofilaments are promising candidates to fulfil this task. We compared two highly sensitive assays to measure two subunits of the neurofilament protein (neurofilament light (NfL) and neurofilament heavy chain (NfH)). METHODS We evaluated the analytical and clinical performance of the UmanDiagnostics NF-light(®) enzyme-linked immunosorbent assay (ELISA) in the cerebrospinal fluid (CSF) of a group of 148 patients with clinically isolated syndrome (CIS) or multiple sclerosis (MS), and 72 controls. We compared our results with referring levels of our previously-developed CSF NfH(SMI35) assay. RESULTS Exposure to room temperature (up to 8 days) or repetitive thawing (up to 4 thaws) did not influence measurement of NfL concentrations. Values of NfL were higher in all disease stages of CIS/MS, in comparison to controls (p ≤ 0.001). NfL levels correlated with the Expanded Disability Status Scale (EDSS) score in patients with relapsing disease (r(s) = 0.31; p = 0.002), spinal cord relapses and with CSF markers of acute inflammation. The ability of NfL to distinguish patients from controls was greater than that of NfH(SMI35) in both CIS patients (p = 0.001) and all MS stages grouped together (p = 0.035). CONCLUSIONS NfL proved to be a stable protein, an important prerequisite for a reliable biomarker, and the NF-light(®) ELISA performed better in discriminating patients from controls, compared with the ECL-NfH(SMI35) immunoassay. We confirmed and expanded upon previous findings regarding neurofilaments as quantitative markers of neurodegeneration. Our results further support the role of neurofilaments as a potential surrogate measure for neuroprotective treatment in MS studies.
Collapse
Affiliation(s)
- Jens Kuhle
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
The diagnostic and prognostic value of neurofilament heavy chain levels in immune-mediated optic neuropathies. Mult Scler Int 2012; 2012:217802. [PMID: 23316360 PMCID: PMC3534363 DOI: 10.1155/2012/217802] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/18/2012] [Accepted: 11/19/2012] [Indexed: 01/03/2023] Open
Abstract
Background. Loss of visual function differs between immune-mediated optic neuropathies and is related to axonal loss in the optic nerve. This study investigated the diagnostic and prognostic value of a biomarker for neurodegeneration, the neurofilament heavy chain (NfH) in three immune-mediated optic neuropathies. Methods. A prospective, longitudinal study including patients with optic neuritis due to multiple sclerosis (MSON, n = 20), chronic relapsing inflammatory optic neuritis (CRION, n = 19), neuromyelitis optica (NMO, n = 9), and healthy controls (n = 28). Serum NfH-SMI35 levels were quantified by ELISA. Findings. Serum NfH-SMI35 levels were highest in patients with NMO (mean 0.79 ± 1.51 ng/mL) compared to patients with CRION (0.13 ± 0.16 ng/mL, P = 0.007), MSON (0.09 ± 0.09, P = 0.008), and healthy controls (0.01 ± 0.02 ng/mL, P = 0.001). High serum NfH-SMI35 levels were related to poor visual outcome. Conclusions. Blood NfH-SMI35 levels are of moderate diagnostic and more important prognostic value in immune-mediated optic neuropathies. We speculate that longitudinal blood NfH levels may help to identify particular disabling events in relapsing conditions.
Collapse
|
47
|
Khalil M, Enzinger C, Langkammer C, Ropele S, Mader A, Trentini A, Vane MLG, Wallner-Blazek M, Bachmaier G, Archelos JJ, Koel-Simmelink MJA, Blankenstein MA, Fuchs S, Fazekas F, Teunissen CE. CSF neurofilament and N-acetylaspartate related brain changes in clinically isolated syndrome. Mult Scler 2012; 19:436-42. [PMID: 22917689 PMCID: PMC3652709 DOI: 10.1177/1352458512458010] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Axonal damage is considered a major cause of disability in multiple sclerosis (MS) and may start early in the disease. Specific biomarkers for this process are of great interest. Objective: To study if cerebrospinal fluid (CSF) biomarkers for axonal damage reflect and predict disease progression already in the earliest stages of the disease, that is, in clinically isolated syndrome (CIS). Methods: We assessed CSF levels of neurofilament heavy (NFH), neurofilament light (NFL) and N-acetylaspartate (NAA) in 67 patients with CIS and 18 controls with neuropsychiatric diseases of non-inflammatory aetiology (NC). Patients with CIS underwent baseline magnetic resonance imaging (MRI) at 3T, and a follow-up MRI after 1 year was obtained in 28 of them. Results: Compared with NC, patients with CIS had higher NFH (p=0.05) and NFL (p<0.001) levels. No significant group differences were found for NAA. Patients’ NFH levels correlated with physical disability (r=0.304, p<0.05) and with change in brain volume over 1 year of follow-up (r=-0.518, p<0.01) but not with change in T2 lesion load. Conclusion: Our results confirm increased neurofilament levels already in CIS being related to the level of physical disability. The association of NFH levels with brain volume but not lesion volume changes supports the association of these markers with axonal damage.
Collapse
Affiliation(s)
- M Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Vetrone SA, Huarng MC, Alocilja EC. Detection of non-PCR amplified S. enteritidis genomic DNA from food matrices using a gold-nanoparticle DNA biosensor: a proof-of-concept study. SENSORS 2012; 12:10487-99. [PMID: 23112611 PMCID: PMC3472839 DOI: 10.3390/s120810487] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/27/2012] [Accepted: 07/31/2012] [Indexed: 01/10/2023]
Abstract
Bacterial pathogens pose an increasing food safety and bioterrorism concern. Current DNA detection methods utilizing sensitive nanotechnology and biosensors have shown excellent detection, but require expensive and time-consuming polymerase chain reaction (PCR) to amplify DNA targets; thus, a faster, more economical method is still essential. In this proof-of-concept study, we investigated the ability of a gold nanoparticle-DNA (AuNP-DNA) biosensor to detect non-PCR amplified genomic Salmonella enterica serovar Enteritidis (S. enteritidis) DNA, from pure or mixed bacterial culture and spiked liquid matrices. Non-PCR amplified DNA was hybridized into sandwich-like structures (magnetic nanoparticles/DNA/AuNPs) and analyzed through detection of gold voltammetric peaks using differential pulse voltammetry. Our preliminary data indicate that non-PCR amplified genomic DNA can be detected at a concentration as low as 100 ng/mL from bacterial cultures and spiked liquid matrices, similar to reported PCR amplified detection levels. These findings also suggest that AuNP-DNA biosensors are a first step towards a viable detection method of bacterial pathogens, in particular, for resource-limited settings, such as field-based or economically limited conditions. Future efforts will focus on further optimization of the DNA extraction method and AuNP-biosensors, to increase sensitivity at lower DNA target concentrations from food matrices comparable to PCR amplified DNA detection strategies.
Collapse
Affiliation(s)
- Sylvia A. Vetrone
- Department of Biology, Whittier College, 13406 E. Philadelphia St., Whittier, CA 90608, USA; E-Mail:
| | - Michael C. Huarng
- Department of Biosystems and Agricultural Engineering, 213 Farrall Hall, Michigan State University, East Lansing, MI 48824, USA
| | - Evangelyn C. Alocilja
- Department of Biosystems and Agricultural Engineering, 213 Farrall Hall, Michigan State University, East Lansing, MI 48824, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-517-355-0083; Fax: +1-517-432-2892
| |
Collapse
|
49
|
Lu CH, Petzold A, Kalmar B, Dick J, Malaspina A, Greensmith L. Plasma neurofilament heavy chain levels correlate to markers of late stage disease progression and treatment response in SOD1(G93A) mice that model ALS. PLoS One 2012; 7:e40998. [PMID: 22815892 PMCID: PMC3397981 DOI: 10.1371/journal.pone.0040998] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 06/15/2012] [Indexed: 12/13/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder characterised by progressive degeneration of motor neurons leading to death, typically within 3–5 years of symptom onset. The diagnosis of ALS is largely reliant on clinical assessment and electrophysiological findings. Neither specific investigative tools nor reliable biomarkers are currently available to enable an early diagnosis or monitoring of disease progression, hindering the design of treatment trials. Methodology/Principal Findings In this study, using the well-established SOD1G93A mouse model of ALS and a new in-house ELISA method, we have validated that plasma neurofilament heavy chain protein (NfH) levels correlate with both functional markers of late stage disease progression and treatment response. We detected a significant increase in plasma levels of phosphorylated NfH during disease progression in SOD1G93A mice from 105 days onwards. Moreover, increased plasma NfH levels correlated with the decline in muscle force, motor unit survival and, more significantly, with the loss of spinal motor neurons in SOD1 mice during this critical period of decline. Importantly, mice treated with the disease modifying compound arimoclomol had lower plasma NfH levels, suggesting plasma NfH levels could be validated as an outcome measure for treatment trials. Conclusions/Significance These results show that plasma NfH levels closely reflect later stages of disease progression and therapeutic response in the SOD1G93A mouse model of ALS and may potentially be a valuable biomarker of later disease progression in ALS.
Collapse
Affiliation(s)
- Ching-Hua Lu
- Sobell Department of Motor Neuroscience and Movement Disorders, MRC Centre for Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, United Kingdom
- Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Axel Petzold
- Department of Neuroinflammation, UCL Institute of Neurology, University College London, London, United Kingdom
- VU Medical Centre, Dept. of Neurology, Amsterdam, The Netherlands
| | - Bernadett Kalmar
- Sobell Department of Motor Neuroscience and Movement Disorders, MRC Centre for Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, United Kingdom
| | - James Dick
- Sobell Department of Motor Neuroscience and Movement Disorders, MRC Centre for Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Andrea Malaspina
- Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- North-East London and Essex MND Care and Research Centre, London, United Kingdom
| | - Linda Greensmith
- Sobell Department of Motor Neuroscience and Movement Disorders, MRC Centre for Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Abstract
Neurodegeneration is the correlate of disease progression in multiple sclerosis (MS) and thus biological biomarkers that sensitively reflect this process are much needed. Neurofilament protein subunits are potential cerebrospinal fluid (CSF) biomarkers for disease progression in MS. We argue that the neurofilament light subunit can reflect acute axonal damage mediated by inflammatory mechanisms and can imply prognostic value for conversion from clinically isolated syndrome (CIS) to definite MS. The neurofilament heavy subunit may rather reflect chronic irreversible damage and has prognostic value for disease progression or disability. The neurofilament intermediate subunit has not yet been studied. Recent studies showing higher neurofilament light or heavy subunit levels to be altered upon treatment regimes indicate their potential clinical value in monitoring treatment or side effects. Future studies should be aimed at the optimisation, standardisation and interlaboratory implementation of the assays and address the predictive value of these biomarkers.
Collapse
|