50 results on '"Claire Bridel"'
Search Results
2. CSF protein ratios with enhanced potential to reflect Alzheimer’s disease pathology and neurodegeneration
- Author
-
Sára Mravinacová, Vilma Alanko, Sofia Bergström, Claire Bridel, Yolande Pijnenburg, Göran Hagman, Miia Kivipelto, Charlotte Teunissen, Peter Nilsson, Anna Matton, and Anna Månberg
- Subjects
Alzheimer’s disease ,CSF ,Neurodegeneration ,Cognitive decline ,Protein ratios ,Protein profiling ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Amyloid and tau aggregates are considered to cause neurodegeneration and consequently cognitive decline in individuals with Alzheimer’s disease (AD). Here, we explore the potential of cerebrospinal fluid (CSF) proteins to reflect AD pathology and cognitive decline, aiming to identify potential biomarkers for monitoring outcomes of disease-modifying therapies targeting these aggregates. Method We used a multiplex antibody-based suspension bead array to measure the levels of 49 proteins in CSF from the Swedish GEDOC memory clinic cohort at the Karolinska University Hospital. The cohort comprised 148 amyloid- and tau-negative individuals (A-T-) and 65 amyloid- and tau-positive individuals (A+T+). An independent sample set of 26 A-T- and 26 A+T+ individuals from the Amsterdam Dementia Cohort was used for validation. The measured proteins were clustered based on their correlation to CSF amyloid beta peptides, tau and NfL levels. Further, we used support vector machine modelling to identify protein pairs, matched based on their cluster origin, that reflect AD pathology and cognitive decline with improved performance compared to single proteins. Results The protein-clustering revealed 11 proteins strongly correlated to t-tau and p-tau (tau-associated group), including mainly synaptic proteins previously found elevated in AD such as NRGN, GAP43 and SNCB. Another 16 proteins showed predominant correlation with Aβ42 (amyloid-associated group), including PTPRN2, NCAN and CHL1. Support vector machine modelling revealed that proteins from the two groups combined in pairs discriminated A-T- from A+T+ individuals with higher accuracy compared to single proteins, as well as compared to protein pairs composed of proteins originating from the same group. Moreover, combining the proteins from different groups in ratios (tau-associated protein/amyloid-associated protein) significantly increased their correlation to cognitive decline measured with cognitive scores. The results were validated in an independent cohort. Conclusions Combining brain-derived proteins in pairs largely enhanced their capacity to discriminate between AD pathology-affected and unaffected individuals and increased their correlation to cognitive decline, potentially due to adjustment of inter-individual variability. With these results, we highlight the potential of protein pairs to monitor neurodegeneration and thereby possibly the efficacy of AD disease-modifying therapies.
- Published
- 2024
- Full Text
- View/download PDF
3. Contrast-Enhancing Lesion Segmentation in Multiple Sclerosis: A Deep Learning Approach Validated in a Multicentric Cohort
- Author
-
Martina Greselin, Po-Jui Lu, Lester Melie-Garcia, Mario Ocampo-Pineda, Riccardo Galbusera, Alessandro Cagol, Matthias Weigel, Nina de Oliveira Siebenborn, Esther Ruberte, Pascal Benkert, Stefanie Müller, Sebastian Finkener, Jochen Vehoff, Giulio Disanto, Oliver Findling, Andrew Chan, Anke Salmen, Caroline Pot, Claire Bridel, Chiara Zecca, Tobias Derfuss, Johanna M. Lieb, Michael Diepers, Franca Wagner, Maria I. Vargas, Renaud Du Pasquier, Patrice H. Lalive, Emanuele Pravatà, Johannes Weber, Claudio Gobbi, David Leppert, Olaf Chan-Hi Kim, Philippe C. Cattin, Robert Hoepner, Patrick Roth, Ludwig Kappos, Jens Kuhle, and Cristina Granziera
- Subjects
deep learning ,multiple sclerosis ,automatic segmentation ,gadolinium contrast-enhancing lesions ,Technology ,Biology (General) ,QH301-705.5 - Abstract
The detection of contrast-enhancing lesions (CELs) is fundamental for the diagnosis and monitoring of patients with multiple sclerosis (MS). This task is time-consuming and suffers from high intra- and inter-rater variability in clinical practice. However, only a few studies proposed automatic approaches for CEL detection. This study aimed to develop a deep learning model that automatically detects and segments CELs in clinical Magnetic Resonance Imaging (MRI) scans. A 3D UNet-based network was trained with clinical MRI from the Swiss Multiple Sclerosis Cohort. The dataset comprised 372 scans from 280 MS patients: 162 showed at least one CEL, while 118 showed no CELs. The input dataset consisted of T1-weighted before and after gadolinium injection, and FLuid Attenuated Inversion Recovery images. The sampling strategy was based on a white matter lesion mask to confirm the existence of real contrast-enhancing lesions. To overcome the dataset imbalance, a weighted loss function was implemented. The Dice Score Coefficient and True Positive and False Positive Rates were 0.76, 0.93, and 0.02, respectively. Based on these results, the model developed in this study might well be considered for clinical decision support.
- Published
- 2024
- Full Text
- View/download PDF
4. Clusters of co-abundant proteins in the brain cortex associated with fronto-temporal lobar degeneration
- Author
-
Claire Bridel, Juami H. M. van Gils, Suzanne S. M. Miedema, Jeroen J. M. Hoozemans, Yolande A. L. Pijnenburg, August B. Smit, Annemieke J. M. Rozemuller, Sanne Abeln, and Charlotte E. Teunissen
- Subjects
Frontotemporal lobar degeneration ,TDP43 ,Tau ,Dementia ,Tissue proteomics ,Co-expression module ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Frontotemporal lobar degeneration (FTLD) is characterized pathologically by neuronal and glial inclusions of hyperphosphorylated tau or by neuronal cytoplasmic inclusions of TDP43. This study aimed at deciphering the molecular mechanisms leading to these distinct pathological subtypes. Methods To this end, we performed an unbiased mass spectrometry-based proteomic and systems-level analysis of the middle frontal gyrus cortices of FTLD-tau (n = 6), FTLD-TDP (n = 15), and control patients (n = 5). We validated these results in an independent patient cohort (total n = 24). Results The middle frontal gyrus cortex proteome was most significantly altered in FTLD-tau compared to controls (294 differentially expressed proteins at FDR = 0.05). The proteomic modifications in FTLD-TDP were more heterogeneous (49 differentially expressed proteins at FDR = 0.1). Weighted co-expression network analysis revealed 17 modules of co-regulated proteins, 13 of which were dysregulated in FTLD-tau. These modules included proteins associated with oxidative phosphorylation, scavenger mechanisms, chromatin regulation, and clathrin-mediated transport in both the frontal and temporal cortex of FTLD-tau. The most strongly dysregulated subnetworks identified cyclin-dependent kinase 5 (CDK5) and polypyrimidine tract-binding protein 1 (PTBP1) as key players in the disease process. Dysregulation of 9 of these modules was confirmed in independent validation data sets of FLTD-tau and control temporal and frontal cortex (total n = 24). Dysregulated modules were primarily associated with changes in astrocyte and endothelial cell protein abundance levels, indicating pathological changes in FTD are not limited to neurons. Conclusions Using this innovative workflow and zooming in on the most strongly dysregulated proteins of the identified modules, we were able to identify disease-associated mechanisms in FTLD-tau with high potential as biomarkers and/or therapeutic targets.
- Published
- 2023
- Full Text
- View/download PDF
5. Paraneoplastic Autoimmune Limbic Encephalitis Associated with an Atypical Carcinoid Tumor of the Lung: A Case Report
- Author
-
Emma Marull Paretas, Claudio De Vito, Sabina Catalano-Chiuve, Maria-Isabel Vargas, Frédéric Assal, Patrice Lalive, and Claire Bridel
- Subjects
cancer ,case report ,cognitive dysfunction ,encephalitis ,magnetic resonance imaging ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
We report the case of a patient with a history of an atypical lung carcinoid tumor who developed a rapidly progressive memory impairment. The clinical presentation as well as brain MRI, cerebrospinal fluid, and laboratory tests led to the diagnosis of seronegative paraneoplastic autoimmune limbic encephalitis. To the best of our knowledge, this is the first case in literature of such association. This case also highlights an exceptionally favorable outcome, both clinically and radiologically, after immunosuppression and tumor removal.
- Published
- 2023
- Full Text
- View/download PDF
6. Identification of novel cerebrospinal fluid biomarker candidates for dementia with Lewy bodies: a proteomic approach
- Author
-
Inger van Steenoven, Marleen J. A. Koel-Simmelink, Leonie J. M. Vergouw, Betty M. Tijms, Sander R. Piersma, Thang V. Pham, Claire Bridel, Gian-Luca Ferri, Cristina Cocco, Barbara Noli, Paul F. Worley, Mei-Fang Xiao, Desheng Xu, Patrick Oeckl, Markus Otto, Wiesje M. van der Flier, Frank Jan de Jong, Connie R. Jimenez, Afina W. Lemstra, and Charlotte E. Teunissen
- Subjects
Biomarkers ,Cerebrospinal fluid ,Dementia with Lewy bodies ,Lewy body dementia ,Proteomics ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Diagnosis of dementia with Lewy bodies (DLB) is challenging, largely due to a lack of diagnostic tools. Cerebrospinal fluid (CSF) biomarkers have been proven useful in Alzheimer’s disease (AD) diagnosis. Here, we aimed to identify novel CSF biomarkers for DLB using a high-throughput proteomic approach. Methods We applied liquid chromatography/tandem mass spectrometry with label-free quantification to identify biomarker candidates to individual CSF samples from a well-characterized cohort comprising patients with DLB (n = 20) and controls (n = 20). Validation was performed using (1) the identical proteomic workflow in an independent cohort (n = 30), (2) proteomic data from patients with related neurodegenerative diseases (n = 149) and (3) orthogonal techniques in an extended cohort consisting of DLB patients and controls (n = 76). Additionally, we utilized random forest analysis to identify the subset of candidate markers that best distinguished DLB from all other groups. Results In total, we identified 1995 proteins. In the discovery cohort, 69 proteins were differentially expressed in DLB compared to controls (p
- Published
- 2020
- Full Text
- View/download PDF
7. Regulator of oligodendrocyte maturation, miR-219, a potential biomarker for MS
- Author
-
Ilona B. Bruinsma, Marie van Dijk, Claire Bridel, Timothy van de Lisdonk, Sanne Q. Haverkort, Tessel F. Runia, Lawrence Steinman, Rogier Q. Hintzen, Joep Killestein, Marcel M. Verbeek, Charlotte E. Teunissen, and Brigit A. de Jong
- Subjects
Multiple sclerosis ,Cerebrospinal fluid ,Biomarkers ,miRNA ,miR-219 ,miR-150 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Multiple sclerosis (MS) is a demyelinating and degenerative disease of the central nervous system. Normally, demyelination is followed by remyelination, which requires repopulation of a demyelinated area by oligodendrocyte precursor cells. Although large numbers of precursor cells are present in MS lesions, remyelination often fails, in part by the inability of precursor cells to differentiate into mature myelin-forming cells. In mouse and rat, miR-219 is required for this differentiation. Previously, we identified decreased miR-219 expression in tissue of MS patients compared to controls. Cell-free miRNAs have been detected in many different body fluids including cerebrospinal fluid (CSF) and may reflect disease processes going on in the central nervous system. This prompted us to investigate the biomarker performance of CSF miR-219 for MS diagnosis. Methods Quantitative PCR was performed measuring miR-219 levels in CSF of MS patients and controls in three independent cohorts. Results All three cohorts of MS patients and controls revealed that absence of miR-219 detection in CSF is consistently associated with MS. Conclusions We have been able to identify and validate absence of miR-219 detection in CSF of MS patients compared to controls, suggesting that it may emerge as a candidate biomarker for MS diagnosis.
- Published
- 2017
- Full Text
- View/download PDF
8. Glutaminyl cyclase activity correlates with levels of Aβ peptides and mediators of angiogenesis in cerebrospinal fluid of Alzheimer’s disease patients
- Author
-
Claire Bridel, Torsten Hoffmann, Antje Meyer, Sisi Durieux, Marleen A. Koel-Simmelink, Matthias Orth, Philip Scheltens, Inge Lues, and Charlotte E. Teunissen
- Subjects
Alzheimer’s disease ,Cerebrospinal fluid ,Glutaminyl cyclase ,Amyloid beta ,3pE-Aβ42 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Pyroglutamylation of truncated Aβ peptides, which is catalysed by enzyme glutaminyl cyclase (QC), generates pE-Aβ species with enhanced aggregation propensities and resistance to most amino-peptidases and endo-peptidases. pE-Aβ species have been identified as major constituents of Aβ plaques and reduction of pE-Aβ species is associated with improvement of cognitive tasks in animal models of Alzheimer’s disease (AD). Pharmacological inhibition of QC has thus emerged as a promising therapeutic approach for AD. Here, we question whether cerebrospinal fluid (CSF) QC enzymatic activity differs between AD patients and controls and whether inflammatory or angiogenesis mediators, some of which are potential QC substrates, and/or Aβ peptides may serve as pharmacodynamic read-outs for QC inhibition. Methods QC activity, Aβ peptides and inflammatory or angiogenesis mediators were measured in CSF of a clinically well-characterized cohort of 20 mild AD patients, 20 moderate AD patients and 20 subjective memory complaints (SMC) controls. Correlation of these parameters with core diagnostic CSF AD biomarkers (Aβ42, tau and p-tau) and clinical features was evaluated. Results QC activity shows a tendency to decrease with AD progression (p = 0.129). The addition of QC activity to biomarkers tau and p-tau significantly increases diagnostic power (ROC-AUCTAU = 0.878, ROC-AUCTAU&QC = 0.939 and ROC-AUCpTAU = 0.820, ROC-AUCpTAU&QC = 0.948). In AD and controls, QC activity correlates with Aβ38 (r = 0.83, p 0.5, p 0.35, p =
- Published
- 2017
- Full Text
- View/download PDF
9. No Plasmatic Proteomic Signature at Clinical Disease Onset Associated With 11 Year Clinical, Cognitive and MRI Outcomes in Relapsing-Remitting Multiple Sclerosis Patients
- Author
-
Claire Bridel, Anand J. C. Eijlers, Wessel N. van Wieringen, Marleen Koel-Simmelink, Cyra E. Leurs, Menno M. Schoonheim, Joep Killestein, and Charlotte E. Teunissen
- Subjects
multiple sclerosis ,prognosis ,proteomics ,MRI ,cognition ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Background: The clinical course of relapsing-remitting multiple sclerosis (RRMS) is highly heterogeneous and prognostic biomarkers at time of diagnosis are lacking.Objective: We investigated the predictive value of the plasma proteome at time of diagnosis in RRMS patients.Methods: The plasma proteome was interrogated using a novel aptamer-based proteomics platform, which allows to measure the levels of a predefined set of 1310 proteins.Results: In 67 clinically and radiologically well characterized RRMS patients, we found no association between the plasma proteome at diagnosis and clinical, cognitive or MRI outcomes after 11 years.Conclusions: Proteomics studies on cerebrospinal fluid may be better suited to identify prognostic biomarkers in early RRMS.
- Published
- 2018
- Full Text
- View/download PDF
10. VGF Peptides in Cerebrospinal Fluid of Patients with Dementia with Lewy Bodies
- Author
-
Inger van Steenoven, Barbara Noli, Cristina Cocco, Gian-Luca Ferri, Patrick Oeckl, Markus Otto, Marleen J. A. Koel-Simmelink, Claire Bridel, Wiesje M. van der Flier, Afina W. Lemstra, and Charlotte E. Teunissen
- Subjects
VGF ,cerebrospinal fluid ,dementia with Lewy bodies ,synaptic dysfunction ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
In a previous proteomic study, we identified the neurosecretory protein VGF (VGF) as a potential biomarker for dementia with Lewy bodies (DLB). Here, we extended the study of VGF by comparing levels in cerebrospinal fluid (CSF) from 44 DLB patients, 20 Alzheimer’s disease (AD) patients, and 22 cognitively normal controls selected from the Amsterdam Dementia Cohort. CSF was analyzed using two orthogonal analytical methods: (1) In-house-developed quantitative ELISA and (2) selected reaction monitoring (SRM). We further addressed associations of VGF with other CSF biomarkers and cognition. VGF levels were lower in CSF from patients with DLB compared to either AD patients or controls. VGF was positively correlated with CSF tau and α-synuclein (0.55 < r < 0.75), but not with Aβ1-42. In DLB patients, low VGF levels were related to a more advanced cognitive decline at time of first presentation, whereas high levels of VGF were associated with steeper subsequent longitudinal cognitive decline. Hence, CSF VGF levels were lower in DLB compared to both AD and controls across different analytical methods. The strong associations with cognitive decline further points out VGF as a possible disease stage or prognostic marker for DLB.
- Published
- 2019
- Full Text
- View/download PDF
11. Correction: Aerosols transmit prions to immunocompetent and immunodeficient mice.
- Author
-
Johannes Haybaeck, Mathias Heikenwalder, Britta Klevenz, Petra Schwarz, Ilan Margalith, Claire Bridel, Kirsten Mertz, Elizabeta Zirdum, Benjamin Petsch, Thomas J Fuchs, Lothar Stitz, and Adriano Aguzzi
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Published
- 2016
- Full Text
- View/download PDF
12. Neurodegeneration and unfolded-protein response in mice expressing a membrane-tethered flexible tail of PrP.
- Author
-
Paolo Dametto, Asvin K K Lakkaraju, Claire Bridel, Lukas Villiger, Tracy O'Connor, Uli S Herrmann, Pawel Pelczar, Thomas Rülicke, Donal McHugh, Arlind Adili, and Adriano Aguzzi
- Subjects
Medicine ,Science - Abstract
The cellular prion protein (PrPC) consists of a flexible N-terminal tail (FT, aa 23-128) hinged to a membrane-anchored globular domain (GD, aa 129-231). Ligation of the GD with antibodies induces rapid neurodegeneration, which is prevented by deletion or functional inactivation of the FT. Therefore, the FT is an allosteric effector of neurotoxicity. To explore its mechanism of action, we generated transgenic mice expressing the FT fused to a GPI anchor, but lacking the GD (PrPΔ141-225, or "FTgpi"). Here we report that FTgpi mice develop a progressive, inexorably lethal neurodegeneration morphologically and biochemically similar to that triggered by anti-GD antibodies. FTgpi was mostly retained in the endoplasmic reticulum, where it triggered a conspicuous unfolded protein response specifically activating the PERK pathway leading to phosphorylation of eIF2α and upregulation of CHOP ultimately leading to neurodegeration similar to what was observed in prion infection.
- Published
- 2015
- Full Text
- View/download PDF
13. Update on multiple sclerosis treatments
- Author
-
Claire Bridel and Patrice H. Lalive
- Subjects
recommendations ,Multiple sclerosis ,disease modifying therapies ,fingolimod ,interferon beta ,glatiramer acetate ,Medicine - Abstract
Relapsing-remitting multiple sclerosis (RRMS) management has dramatically changed over the past decade. New drugs have arrived on the market, allowing for more individualised treatment selection. However, this diversity has increased the complexity of RRMS patient follow-up. In this review, we provide summarised information about treatment efficacy, potential side-effects, follow-up recommendations, vaccinations, and pregnancy safety issues for all currently available disease modifying therapies and those awaiting approval.
- Published
- 2014
- Full Text
- View/download PDF
14. Aerosols transmit prions to immunocompetent and immunodeficient mice.
- Author
-
Johannes Haybaeck, Mathias Heikenwalder, Britta Klevenz, Petra Schwarz, Ilan Margalith, Claire Bridel, Kirsten Mertz, Elizabeta Zirdum, Benjamin Petsch, Thomas J Fuchs, Lothar Stitz, and Adriano Aguzzi
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Prions, the agents causing transmissible spongiform encephalopathies, colonize the brain of hosts after oral, parenteral, intralingual, or even transdermal uptake. However, prions are not generally considered to be airborne. Here we report that inbred and crossbred wild-type mice, as well as tga20 transgenic mice overexpressing PrP(C), efficiently develop scrapie upon exposure to aerosolized prions. NSE-PrP transgenic mice, which express PrP(C) selectively in neurons, were also susceptible to airborne prions. Aerogenic infection occurred also in mice lacking B- and T-lymphocytes, NK-cells, follicular dendritic cells or complement components. Brains of diseased mice contained PrP(Sc) and transmitted scrapie when inoculated into further mice. We conclude that aerogenic exposure to prions is very efficacious and can lead to direct invasion of neural pathways without an obligatory replicative phase in lymphoid organs. This previously unappreciated risk for airborne prion transmission may warrant re-thinking on prion biosafety guidelines in research and diagnostic laboratories.
- Published
- 2011
- Full Text
- View/download PDF
15. Unexpected tolerance of alpha-cleavage of the prion protein to sequence variations.
- Author
-
José B Oliveira-Martins, Sei-ichi Yusa, Anna Maria Calella, Claire Bridel, Frank Baumann, Paolo Dametto, and Adriano Aguzzi
- Subjects
Medicine ,Science - Abstract
The cellular form of the prion protein, PrP(C), undergoes extensive proteolysis at the alpha site (109K [see text]H110). Expression of non-cleavable PrP(C) mutants in transgenic mice correlates with neurotoxicity, suggesting that alpha-cleavage is important for PrP(C) physiology. To gain insights into the mechanisms of alpha-cleavage, we generated a library of PrP(C) mutants with mutations in the region neighbouring the alpha-cleavage site. The prevalence of C1, the carboxy adduct of alpha-cleavage, was determined for each mutant. In cell lines of disparate origin, C1 prevalence was unaffected by variations in charge and hydrophobicity of the region neighbouring the alpha-cleavage site, and by substitutions of the residues in the palindrome that flanks this site. Instead, alpha-cleavage was size-dependently impaired by deletions within the domain 106-119. Almost no cleavage was observed upon full deletion of this domain. These results suggest that alpha-cleavage is executed by an alpha-PrPase whose activity, despite surprisingly limited sequence specificity, is dependent on the size of the central region of PrP(C).
- Published
- 2010
- Full Text
- View/download PDF
16. Serum Glial Fibrillary Acidic Protein Compared With Neurofilament Light Chain as a Biomarker for Disease Progression in Multiple Sclerosis
- Author
-
Stephanie Meier, Eline A.J. Willemse, Sabine Schaedelin, Johanna Oechtering, Johannes Lorscheider, Lester Melie-Garcia, Alessandro Cagol, Muhamed Barakovic, Riccardo Galbusera, Suvitha Subramaniam, Christian Barro, Ahmed Abdelhak, Simon Thebault, Lutz Achtnichts, Patrice Lalive, Stefanie Müller, Caroline Pot, Anke Salmen, Giulio Disanto, Chiara Zecca, Marcus D’Souza, Annette Orleth, Michael Khalil, Arabella Buchmann, Renaud Du Pasquier, Özgür Yaldizli, Tobias Derfuss, Klaus Berger, Marco Hermesdorf, Heinz Wiendl, Fredrik Piehl, Marco Battaglini, Urs Fischer, Ludwig Kappos, Claudio Gobbi, Cristina Granziera, Claire Bridel, David Leppert, Aleksandra Maleska Maceski, Pascal Benkert, Jens Kuhle, and Neurochemistry Laboratory
- Subjects
Neurology (clinical) ,610 Medizin und Gesundheit - Abstract
ImportanceThere is a lack of validated biomarkers for disability progression independent of relapse activity (PIRA) in multiple sclerosis (MS).ObjectiveTo determine how serum glial fibrillary acidic protein (sGFAP) and serum neurofilament light chain (sNfL) correlate with features of disease progression vs acute focal inflammation in MS and how they can prognosticate disease progression.Design, Setting, and ParticipantsData were acquired in the longitudinal Swiss MS cohort (SMSC; a consortium of tertiary referral hospitals) from January 1, 2012, to October 20, 2022. The SMSC is a prospective, multicenter study performed in 8 centers in Switzerland. For this nested study, participants had to meet the following inclusion criteria: cohort 1, patients with MS and either stable or worsening disability and similar baseline Expanded Disability Status Scale scores with no relapses during the entire follow-up; and cohort 2, all SMSC study patients who had initiated and continued B-cell–depleting treatment (ie, ocrelizumab or rituximab).ExposuresPatients received standard immunotherapies or were untreated.Main Outcomes and MeasuresIn cohort 1, sGFAP and sNfL levels were measured longitudinally using Simoa assays. Healthy control samples served as the reference. In cohort 2, sGFAP and sNfL levels were determined cross-sectionally.ResultsThis study included a total of 355 patients (103 [29.0%] in cohort 1: median [IQR] age, 42.1 [33.2-47.6] years; 73 female patients [70.9%]; and 252 [71.0%] in cohort 2: median [IQR] age, 44.3 [33.3-54.7] years; 156 female patients [61.9%]) and 259 healthy controls with a median [IQR] age of 44.3 [36.3-52.3] years and 177 female individuals (68.3%). sGFAP levels in controls increased as a function of age (1.5% per year; P P = .01), and were 14.9% higher in women than in men (P = .004). In cohort 1, patients with worsening progressive MS showed 50.9% higher sGFAP levels compared with those with stable MS after additional sNfL adjustment, whereas the 25% increase of sNfL disappeared after additional sGFAP adjustment. Higher sGFAP at baseline was associated with accelerated gray matter brain volume loss (per doubling: 0.24% per year; P z scores were higher in patients developing future confirmed disability worsening compared with those with stable disability (1.94 [0.36-2.23] vs 0.71 [−0.13 to 1.73]; P = .002); this was not significant for sNfL. However, the combined elevation of z scores of both biomarkers resulted in a 4- to 5-fold increased risk of confirmed disability worsening (hazard ratio [HR], 4.09; 95% CI, 2.04-8.18; P P Conclusions and RelevanceResults of this cohort study suggest that sGFAP is a prognostic biomarker for future PIRA and revealed its complementary potential next to sNfL. sGFAP may serve as a useful biomarker for disease progression in MS in individual patient management and drug development.
- Published
- 2023
17. Serum Glial Fibrillary Acidic Protein compared with Neurofilament Light Chain as Biomarker for Multiple Sclerosis Disease Progression (P5-3.016)
- Author
-
Stephanie Meier, Pascal Benkert, Aleksandra Maleska Maceski, Sabine Schaedelin, Johanna Oechtering, Lester Melie-Garcia, Alessandro Cagol, Muhamed Barakovic, Riccardo Galbusera, Lutz Achtnichts, Patrice H. Lalive, Claire Bridel, Stefanie Müller, Caroline Pot, Anke Salmen, Giulio Disanto, Chiara Zecca, Ahmed Abdelhak, Christian Barro, Simon Thebault, Marcus D’Souza, Tobias Derfuss, Annette Orleth, Michael Khalil, Özguer Yaldizli, Marco Hermesdorf, Heinz Wiendl, Fredrik Piehl, Urs Fischer, Ludwig Kappos, Claudio Gobbi, Cristina Granziera, David Leppert, Eline A.J. Willemse, and Jens Kuhle
- Published
- 2023
18. A bilateral aseptic pyogenic ventriculitis following a course of pembrolizumab, an anti-PD-1 immune checkpoint inhibitor treatment for metastatic small cell lung cancer
- Author
-
Jean Terrier, Alan Relecom, Simon Borgeaud, Claire Bridel, Jörg Seebach, Frédéric Assal, Jean-Luc Reny, and Jacques Serratrice
- Subjects
Pharmacology (medical) - Published
- 2022
19. Associating Alzheimer’s disease pathology with its cerebrospinal fluid biomarkers
- Author
-
Claire Bridel, Charisse Somers, Anne Sieben, Annemieke Rozemuller, Ellis Niemantsverdriet, Hanne Struyfs, Yannick Vermeiren, Christine Van Broeckhoven, Peter P De Deyn, Maria Bjerke, Guy Nagels, Charlotte E Teunissen, Sebastiaan Engelborghs, Pathologic Biochemistry and Physiology, Clinical Biology, Clinical sciences, Neuroprotection & Neuromodulation, Artificial Intelligence supported Modelling in clinical Sciences, Neurology, Pathology, Amsterdam Neuroscience - Neurodegeneration, Laboratory Medicine, and Amsterdam Neuroscience - Neuroinfection & -inflammation
- Subjects
Male ,Threonine ,Amyloid beta-Peptides ,Neuroscience(all) ,Apolipoprotein E4 ,tau Proteins ,Plaque, Amyloid ,cerebrospinal fluid ,Peptide Fragments ,Nutritional Biology ,Alzheimer Disease ,Montine criteria ,Disease Progression ,Life Science ,Humans ,Female ,Human medicine ,Neurology (clinical) ,NEUROPATHOLOGY ,Biology ,Alzheimer’s disease ,Biomarkers ,Aged - Abstract
Alzheimer’s disease CSF biomarkers 42 amino acid long amyloid-β peptide (Aβ1–42), total tau protein (T-tau), and tau protein phosphorylated at threonine 181 (P-tau181) are considered surrogate biomarkers of Alzheimer’s disease pathology, and significantly improve diagnostic accuracy. Their ability to reflect neuropathological changes later in the disease course is not well characterized. This study aimed to assess the potential of CSF biomarkers measured in mid to late stage Alzheimer’s disease to reflect post-mortem neuropathological changes. Individuals were selected from two autopsy cohorts of Alzheimer’s disease patients in Antwerp and Amsterdam. Neuropathological diagnosis was performed according to the updated consensus National Institute on Aging-Alzheimer’s Association guidelines, which includes quantification of amyloid-β plaque, neurofibrillary tangle, and neuritic plaque load. CSF samples were analysed for Aβ1–42, T-tau, and P-tau181 by ELISA. One hundred and fourteen cases of pure definite Alzheimer’s disease were included in the study (mean age 74 years, disease duration 6 years at CSF sampling, 50% females). Median interval between CSF sampling and death was 1 year. We found no association between Aβ1–42 and Alzheimer’s disease neuropathological change profile. In contrast, an association of P-tau181 and T-tau with Alzheimer’s disease neuropathological change profile was observed. P-tau181 was associated with all three individual Montine scores, and the associations became stronger and more significant as the interval between lumbar puncture and death increased. T-tau was also associated with all three Montine scores, but in individuals with longer intervals from lumbar puncture to death only. Stratification of the cohort according to APOE ε4 carrier status revealed that the associations applied mostly to APOE ε4 non-carriers. Our data suggest that similar to what has been reported for Aβ1–42, plateau levels of P-tau181 and T-tau are reached during the disease course, albeit at later disease stages, reducing the potential of tau biomarkers to monitor Alzheimer’s disease pathology as the disease progresses. As a consequence, CSF biomarkers, which are performant for clinical diagnosis of early Alzheimer’s disease, may not be well suited for staging or monitoring Alzheimer’s disease pathology as it progresses through later stages.
- Published
- 2022
20. Clusters of co-regulated proteins in brain cortex associate with fronto-temporal lobar degeneration
- Author
-
Claire Bridel, Juami HM van Gils, Suzanne SM Miedema, Jeroen J Hoozemans, Yolande AL Pijnenburg, August B Smit, Annemieke JM Rozemuller, Sanne Abeln, and Charlotte E Teunissen
- Abstract
Background Frontotemporal lobar degeneration (FTLD) is characterized pathologically by neuronal and glial inclusions of hyperphosphorylated tau or by neuronal cytoplasmic inclusions of TDP43. This study aimed at deciphering the molecular mechanisms leading to these distinct pathological subtypes. Methods To this end, we performed an unbiased mass spectrometry-based proteomic and systems-level analysis of middle frontal gyrus cortices of FTLD-tau (n = 6), FTLD-TDP (n = 15), and control patients (n = 5). We validated these results in an independent patient cohort (total n = 24). Results The middle frontal gyrus cortex proteome was most significantly altered in FTLD-tau compared to controls (294 differentially expressed proteins at FDR = 0.05). The proteomic modifications in FTLD-TDP were more heterogeneous (49 differentially expressed proteins at FDR = 0.1). Weighted co-expression network analysis revealed 17 modules of co-regulated proteins, 13 of which were dysregulated in FTLD-tau. These modules included proteins associated with oxidative phosphorylation, scavenger mechanisms, chromatin regulation and clathrin-mediated transport in both the frontal and temporal cortex of FTLD-tau. The most strongly dysregulated subnetworks identified Cyclin-Dependent Kinase 5 (CDK5) and Polypyrimidine Tract Binding Protein 1 (PTBP1) as key players in the disease process. Dysregulation of 9 of these modules was confirmed in independent validation datasets of FLTD-tau and control temporal and frontal cortex (total n = 24). Dysregulated modules were primarily associated with changes in astrocyte and endothelial cell protein expression levels, indicating pathological changes in FTD are not limited to neurons. Conclusions Using this innovative workflow and zooming in on the most strongly dysregulated proteins of the identified modules, we were able to identify disease-associated mechanisms in FTLD-tau with high potential as biomarkers and/or therapeutic targets.
- Published
- 2022
21. Encéphalopathie aiguë et Covid-19: hypothèses physiopathologiques
- Author
-
Marjolaine Uginet, Patrick Stancu, Claire Bridel, Jacques Serratrice, Patrice H. Lalive, and Matteo Coen
- Subjects
General Medicine - Published
- 2021
22. Blood-based protein biomarkers in definite frontotemporal dementia: a case-control study
- Author
-
Charlotte Teunissen, Marta Del Campo, Carel Peeters, Lieke Meeter, Harro Seelaar, Marleen Koel-Simmelink, Inez Ramakers, H Middelkoop, Peter De Deyn, Jurgen Claessen, John van Swieten, Claire Bridel, Jeroen Hoozemans, Wiesje van der Flier, Philip Scheltens, Yolande Pijnenburg, and Rosha Babapour-Mofrad
- Abstract
Background: Frontotemporal dementia (FTD) is caused by frontotemporal lobar degeneration (FTLD), characterized mainly by inclusions of Tau (FTLD-Tau) or TAR DNA binding43 (FTLD-TDP) proteins. Plasma biomarkers are strongly needed for specific diagnosis and potential treatment monitoring of FTD. We aimed to identify specific FTD plasma biomarker profiles discriminating FTD from AD and controls, and between FTD pathological subtypes. In addition, we compared plasma results with results in post-mortem frontal cortex of FTD cases to understand the underlying process. Methods: Plasma proteins (n=1303) from pathologically and/or genetically confirmed FTD patients (n=56; FTLD-Tau n=16; age= 58.2±6.2; 44% female, FTLD-TDP n= 40; age= 59.8±7.9; 45% female), AD patients (n=57; age= 65.5±8.0; 39% female), and non-demented controls (n=148; 61.3±7.9; 41% female) were measured using an aptamer-based proteomic technology (SomaScan). In addition, tissue proteins of post-mortem frontal brain cortex of FTD (n=10; FTLD-Tau n=5; age= 56.2±6.9, 60% female, and FTLD-TDP n= 5; age= 64.0±7.7, 60% female) and non-demented controls (n=5; age= 61.3±8.1; 75% female) were measured. Differentially regulated plasma and tissue proteins were identified by global testing adjusting for demographic variables and multiple testing. Logistic lasso regression was used to identify plasma protein panels discriminating FTD from non-demented controls and AD, or FTLD-Tau from FTLD-TDP. Performance of the discriminatory plasma protein panels was based on predictions obtained from bootstrapping with 1000 resampled analysis. Results: Overall plasma protein expression profiles differed between FTD, AD and controls (6 proteins; p=0.005). The overall tissue protein expression profile differed between FTD and controls (7-proteins; p=0.003). There was no difference in overall plasma or tissue expression profile between FTD subtypes. Regression analysis revealed a panel of 12-plasma proteins discriminating FTD from AD with high accuracy (AUC: 0.99). No plasma protein panels discriminating FTD from controls or FTD pathological subtypes were identified. Conclusions: We identified a promising plasma protein panel as a minimally-invasive tool to aid in the differential diagnosis of FTD from AD, which were mainly associated to AD pathophysiology. The lack of plasma profiles specifically associated to FTD or its pathological subtypes might be explained by FTD heterogeneity, calling for FTD studies using large and well-characterize cohorts.
- Published
- 2022
23. Serum Neurofilament Light Association With Progression in Natalizumab-Treated Patients With Relapsing-Remitting Multiple Sclerosis
- Author
-
Iris Dekker, B. Moraal, Joep Killestein, Claire Bridel, Zoë Y.G.J. van Lierop, Frederik Barkhof, Harry Twaalfhoven, Cyra E. Leurs, Charlotte E. Teunissen, Zoé L.E. van Kempen, Bernard M. J. Uitdehaag, Clinical chemistry, Neurology, Amsterdam Neuroscience - Neuroinfection & -inflammation, Radiology and nuclear medicine, Amsterdam Neuroscience - Brain Imaging, AII - Inflammatory diseases, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
medicine.medical_specialty ,Multiple Sclerosis ,Expanded Disability Status Scale ,Adolescent ,business.industry ,Natalizumab ,Neurofilament light ,Multiple sclerosis ,Intermediate Filaments ,medicine.disease ,Multiple Sclerosis, Relapsing-Remitting ,Relapsing remitting ,Interquartile range ,Internal medicine ,Cohort ,Disease Progression ,medicine ,Humans ,Immunologic Factors ,Prospective Studies ,Neurology (clinical) ,business ,medicine.drug ,Cohort study - Abstract
Background and ObjectivesTo investigate the potential of serum neurofilament light (NfL) to reflect or predict progression mostly independent of acute inflammatory disease activity in patients with relapsing-remitting multiple sclerosis (RRMS) treated with natalizumab.MethodsPatients were selected from a prospective observational cohort study initiated in 2006 at the VU University Medical Center Amsterdam, the Netherlands, including patients with RRMS treated with natalizumab. Selection criteria included an age of 18 years or older and a minimum follow-up of 3 years from natalizumab initiation. Clinical and MRI assessments were performed on a yearly basis, and serum NfL was measured at 5 time points during the follow-up, including on the day of natalizumab initiation (baseline), 3 months, 1 year, and 2 years after natalizumab initiation, and on last follow-up visit. Using general linear regression models, we compared the longitudinal dynamics of NfL between patients with and without confirmed Expanded Disability Status Scale (EDSS) progression between year 1 visit and last follow-up, and between individuals with and without EDSS+ progression, a composite endpoint including the EDSS, 9-hole peg test, and timed 25-foot walk.ResultsEighty-nine natalizumab-treated patients with RRMS were included. Median follow-up time was 5.2 years (interquartile range [IQR] 4.3–6.7, range 3.0–11.0) after natalizumab initiation, mean age at time of natalizumab initiation was 36.9 years (SD 8.5), and median disease duration was 7.4 years (IQR 3.8–12.1). Between year 1 and the last follow-up, 28/89 (31.5%) individuals showed confirmed EDSS progression. Data for the EDSS+ endpoint was available for 73 out of the 89 patients and 35/73 (47.9%) showed confirmed EDSS+ progression. We observed a significant reduction in NfL levels 3 months after natalizumab initiation, which reached its nadir of close to 50% of baseline levels 1 year after treatment initiation. We found no difference in the longitudinal dynamics of NfL in progressors vs nonprogressors. NfL levels at baseline and 1 year after natalizumab initiation did not predict progression at last follow-up.ConclusionIn our cohort of natalizumab-treated patients with RRMS, NfL fails to capture or predict progression that occurs largely independently of clinical or radiologic signs of acute focal inflammatory disease activity. Additional biomarkers may thus be needed to monitor progression in these patients.Classification of EvidenceThis study provides Class II evidence that serum NfL levels are not associated with disease progression in natalizumab-treated patients with RRMS.
- Published
- 2021
24. Association of Brain Atrophy With Disease Progression Independent of Relapse Activity in Patients With Relapsing Multiple Sclerosis
- Author
-
Alessandro Cagol, Sabine Schaedelin, Muhamed Barakovic, Pascal Benkert, Ramona-Alexandra Todea, Reza Rahmanzadeh, Riccardo Galbusera, Po-Jui Lu, Matthias Weigel, Lester Melie-Garcia, Esther Ruberte, Nina Siebenborn, Marco Battaglini, Ernst-Wilhelm Radue, Özgür Yaldizli, Johanna Oechtering, Tim Sinnecker, Johannes Lorscheider, Bettina Fischer-Barnicol, Stefanie Müller, Lutz Achtnichts, Jochen Vehoff, Giulio Disanto, Oliver Findling, Andrew Chan, Anke Salmen, Caroline Pot, Claire Bridel, Chiara Zecca, Tobias Derfuss, Johanna M. Lieb, Luca Remonda, Franca Wagner, Maria I. Vargas, Renaud Du Pasquier, Patrice H. Lalive, Emanuele Pravatà, Johannes Weber, Philippe C. Cattin, Claudio Gobbi, David Leppert, Ludwig Kappos, Jens Kuhle, Cristina Granziera, and Clinical chemistry
- Subjects
Adult ,Male ,Multiple Sclerosis ,Brain ,Neurodegenerative Diseases ,Nervous System Malformations ,Magnetic Resonance Imaging ,Disability Evaluation ,Multiple Sclerosis, Relapsing-Remitting ,Central Nervous System Diseases ,Recurrence ,Disease Progression ,Humans ,Female ,Longitudinal Studies ,Neurology (clinical) ,Atrophy ,Gray Matter ,610 Medicine & health - Abstract
Importance The mechanisms driving neurodegeneration and brain atrophy in relapsing multiple sclerosis (RMS) are not completely understood. Objective To determine whether disability progression independent of relapse activity (PIRA) in patients with RMS is associated with accelerated brain tissue loss. Design, Setting, and Participants In this observational, longitudinal cohort study with median (IQR) follow-up of 3.2 years (2.0-4.9), data were acquired from January 2012 to September 2019 in a consortium of tertiary university and nonuniversity referral hospitals. Patients were included if they had regular clinical follow-up and at least 2 brain magnetic resonance imaging (MRI) scans suitable for volumetric analysis. Data were analyzed between January 2020 and March 2021. Exposures According to the clinical evolution during the entire observation, patients were classified as those presenting (1) relapse activity only, (2) PIRA episodes only, (3) mixed activity, or (4) clinical stability. Main Outcomes and Measures Mean difference in annual percentage change (MD-APC) in brain volume/cortical thickness between groups, calculated after propensity score matching. Brain atrophy rates, and their association with the variables of interest, were explored with linear mixed-effect models. Results Included were 1904 brain MRI scans from 516 patients with RMS (67.4% female; mean [SD] age, 41.4 [11.1] years; median [IQR] Expanded Disability Status Scale score, 2.0 [1.5-3.0]). Scans with insufficient quality were excluded (n = 19). Radiological inflammatory activity was associated with increased atrophy rates in several brain compartments, while an increased annualized relapse rate was linked to accelerated deep gray matter (GM) volume loss. When compared with clinically stable patients, patients with PIRA had an increased rate of brain volume loss (MD-APC, -0.36; 95% CI, -0.60 to -0.12; P = .02), mainly driven by GM loss in the cerebral cortex. Patients who were relapsing presented increased whole brain atrophy (MD-APC, -0.18; 95% CI, -0.34 to -0.02; P = .04) with respect to clinically stable patients, with accelerated GM loss in both cerebral cortex and deep GM. No differences in brain atrophy rates were measured between patients with PIRA and those presenting relapse activity. Conclusions and Relevance Our study shows that patients with RMS and PIRA exhibit accelerated brain atrophy, especially in the cerebral cortex. These results point to the need to recognize the insidious manifestations of PIRA in clinical practice and to further evaluate treatment strategies for patients with PIRA in clinical trials.
- Published
- 2022
25. [COVID-19-related acute encephalopathy: physiopathological hypothesis]
- Author
-
Marjolaine, Uginet, Patrick, Stancu, Claire, Bridel, Jacques, Serratrice, Patrice H, Lalive, and Matteo, Coen
- Subjects
Brain Diseases ,Blood-Brain Barrier ,SARS-CoV-2 ,COVID-19 ,Humans ,Nervous System Diseases - Abstract
Acute encephalopathy is one of the most frequent neurological complication in patients hospitalized for COVID-19. Electrolyte imbalance, drugs, and hypoxemia can all affect brain homeostasis, leading to acute cognitive dysfunction and direct implications of the SARS-CoV-2 are not completely understood. Neurological complications of SARS-CoV-2 infection are poorly understood: an inflammatory insult to the endothelium affecting the blood-brain barrier may explain the clinical presentation, but other hypotheses including direct viral damage or an immune-mediated reaction are also suggested. Among these various potential mechanisms, often combined, the controversy remains.L’encéphalopathie aiguë est l’une des principales complications neurologiques des patients hospitalisés pour un Covid-19. Si les complications électrolytiques, les différents traitements et l’hypoxémie ont un effet sur l’homéostasie cérébrale entraînant une perturbation de la cognition, les conséquences cérébrales directes ou indirectes du SARS-CoV-2 ne sont pas complètement élucidées. L’implication du virus entraînant une atteinte endothéliale des vaisseaux cérébraux pourrait participer à cette encéphalopathie, via une fragilisation de la barrière hémato-encéphalique et un passage facilité des cytokines pro-inflammatoires. L’atteinte virale directe ou la réaction autoimmune secondaire ont également été invoquées. Face aux multiples mécanismes physiopathologiques possibles pouvant expliquer cette encéphalopathie, le débat est ouvert.
- Published
- 2021
26. Variations in consecutive serum neurofilament light levels in healthy controls and multiple sclerosis patients
- Author
-
Charlotte E. Teunissen, J.J.A. Heijst, Inge M.W. Verberk, Joep Killestein, Claire Bridel, Clinical chemistry, Amsterdam Neuroscience - Neurodegeneration, Neurology, AII - Inflammatory diseases, Amsterdam Neuroscience - Neuroinfection & -inflammation, and Amsterdam Reproduction & Development (AR&D)
- Subjects
medicine.medical_specialty ,Neurology ,Multiple Sclerosis ,Neuronal protein ,Neurofilament light ,Intermediate Filaments ,Gastroenterology ,Primary progressive ,03 medical and health sciences ,0302 clinical medicine ,Multiple Sclerosis, Relapsing-Remitting ,Neurofilament Proteins ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Secondary progressive ,business.industry ,Multiple sclerosis ,General Medicine ,medicine.disease ,Cohort ,Biomarker (medicine) ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Background Neurofilament light is a neuronal protein detectable in serum (sNfL), with high potential as disease activity biomarker in multiple sclerosis (MS). To date, little is known about sNfL fluctuations between 2 consecutive measurements in healthy controls (HC) and MS patients. Yet this information is critical, as it will help define a clinically significant variation. Methods sNfL was measured at 2 consecutive time points in a cohort of 90 MS patients (untreated relapsing remitting MS (uRRMS), n=35; treated relapsing remitting MS (tRRMS), n= 21; secondary progressive MS, SPMS, n=21; primary progressive MS, PPMS, n=13), and 90 age-matched HC, using the Simoa NfL light® assay. Results Mean sNfL was elevated in all MS subtypes compared to HC (p
- Published
- 2021
27. Author Response: Serum Neurofilament Light Association With Progression in Natalizumab-Treated Patients With Relapsing-Remitting Multiple Sclerosis
- Author
-
Charlotte Teunissen, Claire Bridel, Joep Killestein, Clinical chemistry, Neurology, AII - Inflammatory diseases, Amsterdam Neuroscience - Neuroinfection & -inflammation, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
Neurology (clinical) - Published
- 2022
28. Brain endothelial cell expression of SPARCL-1 is specific to chronic multiple sclerosis lesions and is regulated by inflammatory mediators in vitro
- Author
-
Hugo Vrenken, H.E. de Vries, Connie R. Jimenez, P Gami, Charlotte E. Teunissen, Sander R. Piersma, Jens Kuhle, Erik Nutma, Ellen Iacobaeus, S Durieux, R Gorter, C Derada Troletti, Lou Brundin, Joep Killestein, Laura A. N. Peferoen, Marleen J.A. Koel-Simmelink, Claire Bridel, Sandra Amor, Thang V. Pham, Clinical chemistry, Pathology, Amsterdam Neuroscience - Neuroinfection & -inflammation, Molecular cell biology and Immunology, Radiology and nuclear medicine, Neurology, AII - Inflammatory diseases, Medical oncology laboratory, ACS - Microcirculation, and AGEM - Re-generation and cancer of the digestive system
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Multiple Sclerosis ,Histology ,Neuropathology ,Blood–brain barrier ,Pathology and Forensic Medicine ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Physiology (medical) ,Humans ,Medicine ,Extracellular Matrix Proteins ,business.industry ,Multiple sclerosis ,Calcium-Binding Proteins ,Brain ,Endothelial Cells ,Human brain ,Middle Aged ,medicine.disease ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Immunology ,Disease Progression ,Biomarker (medicine) ,Female ,Endothelium, Vascular ,Neurology (clinical) ,Inflammation Mediators ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Aims Cell matrix modulating protein SPARCL-1 is highly expressed by astrocytes during CNS development and following acute CNS damage. Applying NanoLC-MS/MS to CSF of RRMS and SPMS patients, we identified SPARCL-1 as differentially expressed between these two stages of MS, suggesting a potential as CSF biomarker to differentiate RRMS from SPMS and a role in MS pathogenesis. Methods This study examines the potential of SPARCL-1 as CSF biomarker discriminating RRMS from SPMS in 3 independent cohorts (n=249), analyses its expression pattern in MS lesions (n= 26), and studies its regulation in cultured human brain microvasculature endothelial cells (BEC) after exposure to MS-relevant inflammatory mediators. Results SPARCL-1 expression in CSF was significantly higher in SPMS compared to RRMS in a Dutch cohort of 76 patients. This finding was not replicated in 2 additional cohorts of MS patients from Sweden (n=81) and Switzerland (n=92). In chronic MS lesions, but not active lesions or NAWM, a vessel expression pattern of SPARCL-1 was observed in addition to the expression by astrocytes. EC were found to express SPARCL-1 in chronic MS lesions, and SPARCL-1 expression was regulated by MS-relevant inflammatory mediators in cultured human BEC. Conclusions Conflicting results of SPARCL-1's differential expression in CSF of 3 independent cohorts of RRMS and SPMS patients precludes its use as biomarker for disease progression. The expression of SPARCL-1 by BEC in chronic MS lesions together with its regulation by inflammatory mediators in vitro suggest a role for SPARCL-1 in MS neuropathology, possibly at the brain vascular level. This article is protected by copyright. All rights reserved.
- Published
- 2018
29. Pre-analytical stability of novel cerebrospinal fluid biomarkers
- Author
-
Claire Bridel, Vera M. Mendes, Naomi De Roeck, Ulf Andreasson, Erwin E. W. Jansen, María-Salud García-Ayllón, Eline A.J. Willemse, Peter Paul De Deyn, Charlotte E. Teunissen, Eduard A. Struys, Yannick Vermeiren, Bruno Manadas, Eugeen Vanmechelen, Wiesje M. van der Flier, Javier Sáez-Valero, Inmaculada B. Lopez-Font, Sebastiaan Engelborghs, Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Clinical chemistry, Amsterdam Neuroscience - Neurodegeneration, APH - Personalized Medicine, APH - Methodology, AGEM - Endocrinology, metabolism and nutrition, AGEM - Inborn errors of metabolism, Molecular Neuroscience and Ageing Research (MOLAR), Netherlands Organization for Scientific Research, European Commission, Fundação para a Ciência e a Tecnologia (Portugal), Clinical sciences, and Neurology
- Subjects
0301 basic medicine ,YKL-40 ,Aché ,Assay validation ,Clinical Biochemistry ,Human cerebrospinal fluid ,PROTEIN ,Enzyme-Linked Immunosorbent Assay ,ISSUE ,Biochemistry ,Assay validation, Biomarkers, Human cerebrospinal fluid, Neurodegenerative diseases, Pre-analytical stability ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cerebrospinal fluid ,medicine ,Amyloid precursor protein ,Humans ,Neurogranin ,BRAIN ,Biology ,Theobromine ,Medicine(all) ,chemistry.chemical_classification ,PLASMA ,biology ,Chemistry ,DEMENTIA ,Biochemistry (medical) ,Homovanillic acid ,Neurodegenerative diseases ,General Medicine ,Molecular biology ,Acetylcholinesterase ,language.human_language ,NEUROGRANIN ,ALZHEIMERS-DISEASE ,030104 developmental biology ,Enzyme ,Pre-analytical stability ,030220 oncology & carcinogenesis ,language ,biology.protein ,Human medicine ,Nervous System Diseases ,COLLECTION ,Biomarkers ,MONOAMINE METABOLITES ,medicine.drug - Abstract
Stability of the cerebrospinal fluid (CSF) composition under different pre-analytical conditions is relevant for the diagnostic potential of biomarkers. Our aim was to examine the pre-analytical stability of promising CSF biomarkers that are currently evaluated for their discriminative use in various neurological diseases. Pooled CSF was aliquoted and experimentally exposed to delayed storage: 0, 1, 2, 4, 24, 72, or 168 h at 4 °C or room temperature (RT), or 1–4 months at −20 °C; or up to 7 freeze/thaw (f/t) cycles, before final storage at −80 °C. Eleven CSF biomarkers were screened using immunoassays, liquid chromatography, or enzymatic methods. Levels of neurogranin (truncP75), chitinase-3-like protein (YKL-40), beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), acetylcholinesterase (AChE) enzymatic activity, theobromine, secreted protein acidic and rich in cysteine-like 1 (SPARCL-1) and homovanillic acid (HVA) levels were not affected by the applied storage conditions. 3-Methoxy-4-hydroxyphenylglycol (MHPG) levels linearly and strongly decreased after 4 h at RT (−10%) or 24 h at 4 °C (−27%), and with 6% after every f/t cycle. 5-Methyltetrahydrofolate (5-MTHF) (−29% after 1 week at RT) and 5-hydroxyindoleacetic acid levels (5-HIAA) (−16% after 1 week at RT) were reduced and 3,4-dihydroxyphenylacetic acid (DOPAC) levels (+22% after 1 week at RT) increased, but only after >24 h at RT. Ten out of eleven potential CSF novel biomarkers showed very limited change under common storage and f/t conditions, suggesting that these CSF biomarkers can be trustfully tested under the pre-analytical conditions present across different cohorts., This research was financially supported by Biobanking and BioMolecular resources Research Infrastructure the Netherlands (BBMRI-NL), a research infrastructure financed by the Dutch Government (NWO 184.021.007) under project CP2013-68. This study commenced within the BIOMARKAPD program of the EU Joint Programme – Neurodegenerative Disease Research (JPND). Project supported by the European Regional Development Fund (ERDF) through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT – Fundação para a Ciência e a Tecnologia, I.P., under the projects POCI-01-0145-FEDER-007440, POCI-01-0145-FEDER-016428, and POCI-01-0145-FEDER-016795, and by The National Mass Spectrometry Network (RNEM) under the contract POCI-01-0145-FEDER-402-022125.
- Published
- 2019
30. Diagnostic Value of Cerebrospinal Fluid Neurofilament Light Protein in Neurology: A Systematic Review and Meta-analysis
- Author
-
van Wieringen Wn, Marcel M. Verbeek, Ludwig Kappos, van Swieten Jc, Tove Christensen, Edward J. Wild, Lieke H.H. Meeter, Mattias Vågberg, Ross W. Paterson, Tobias Skillbäck, Lu Ch, Markus Axelsson, Shorena Janelidze, Ulf Andreasson, Maria Bjerke, Jonathan M. Schott, José C. Álvarez-Cermeño, Megan K. Herbert, Nadia K. Magdalinou, Michael Jonsson, Betty M. Tijms, Peter Sundström, Troiano M, Fredrik Piehl, Mohsen Khademi, Pyykkö Ot, Rosanna Tortelli, Jens Kuhle, Lena Brundin, Ales Bartos, Joel Jakobsson, Jessen-Krut J, Michael Khalil, Isabella Laura Simone, Stilund M, Julio C. Rojas, Carole Scherling, Lenka Fialová, David Bäckström, Finn Sellebjerg, Anders Wallin, Jette L. Frederiksen, Pieter Jelle Visser, Signe Modvig, Henrik Zetterberg, Mikael Landén, Mehta, Carla Tortorella, Gudmundur Johannsson, Andrea Malaspina, Giancarlo Logroscino, Pijnenburg Yal, Pérez-Santiago J, Claire Bridel, Weiss A, Romme Christensen J, Niklas Mattsson, Martin Gunnarsson, Alessandro Trentini, Sandberg L, Sara Hall, Kaj Blennow, Lars Forsgren, Ragnarsson O, Oskar Hansson, Jan Lycke, Tomas Olsson, Magnus Gisslén, Joachim Burman, Carsten Wikkelsö, Anders Svenningsson, Luisa M. Villar, Leinonen, Martin R Turner, Charlotte E. Teunissen, Elizabeth Gray, A. Boxer, Neurology, Human genetics, Laboratory Medicine, Epidemiology and Data Science, Amsterdam Neuroscience - Neuroinfection & -inflammation, CCA - Imaging and biomarkers, Clinical sciences, and Group, NFL
- Subjects
NATIONAL INSTITUTE ,medicine.medical_specialty ,Neurology ,neuroaxonal damage ,CLINICAL-DIAGNOSIS ,cerebrospinal fluid ,AMYOTROPHIC-LATERAL-SCLEROSIS ,NO ,AXONAL DAMAGE ,03 medical and health sciences ,0302 clinical medicine ,PARKINSONS-DISEASE ,Internal medicine ,medicine ,Dementia ,030212 general & internal medicine ,Amyotrophic lateral sclerosis ,FIBRILLARY ACIDIC PROTEIN ,Original Investigation ,Medicine(all) ,Neurofilament light protein ,business.industry ,Multiple sclerosis ,MULTIPLE-SCLEROSIS ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,Amyotrophic-lateral-sclerosis ,fibrillary acidic protein ,csf neurofilament ,multiple-sclerosis ,alzheimers-disease ,axonal damage ,neurodegenerative diseases ,parkinsons-disease ,clinical-diagnosis ,national institute ,3. Good health ,ALZHEIMERS-DISEASE ,CSF NEUROFILAMENT ,Meta-analysis ,Biomarker (medicine) ,healthy controls ,Neurology (clinical) ,diagnostic value ,Alzheimer's disease ,business ,NEURODEGENERATIVE DISEASES ,030217 neurology & neurosurgery ,Frontotemporal dementia - Abstract
Key PointsQuestionHow do levels of neurofilament light in cerebrospinal fluid (cNfL) compare between neurological conditions and with healthy controls? FindingsAmong 10 059 individuals in this systematic review and meta-analysis, cNfL was elevated in most neurological conditions compared with healthy controls, and the magnitude of the increase varies extensively. Although cNfL overlaps between most clinically similar conditions, its distribution did not overlap in frontotemporal dementia and other dementias or in Parkinson disease and atypical parkinsonian syndromes. MeaningThe cNfL is a marker of neuronal damage and may be useful to differentiate some clinically similar conditions, such as frontotemporal dementia from Alzheimer disease and Parkinson disease from atypical parkinsonian syndromes. This systematic review and meta-analysis assesses the associations of age, sex, and diagnosis with neurofilament light in cerebrospinal fluid and evaluates its potential in discriminating clinically similar conditions. ImportanceNeurofilament light protein (NfL) is elevated in cerebrospinal fluid (CSF) of a number of neurological conditions compared with healthy controls (HC) and is a candidate biomarker for neuroaxonal damage. The influence of age and sex is largely unknown, and levels across neurological disorders have not been compared systematically to date. ObjectivesTo assess the associations of age, sex, and diagnosis with NfL in CSF (cNfL) and to evaluate its potential in discriminating clinically similar conditions. Data SourcesPubMed was searched for studies published between January 1, 2006, and January 1, 2016, reporting cNfL levels (using the search terms neurofilament light and cerebrospinal fluid) in neurological or psychiatric conditions and/or in HC. Study SelectionStudies reporting NfL levels measured in lumbar CSF using a commercially available immunoassay, as well as age and sex. Data Extraction and SynthesisIndividual-level data were requested from study authors. Generalized linear mixed-effects models were used to estimate the fixed effects of age, sex, and diagnosis on log-transformed NfL levels, with cohort of origin modeled as a random intercept. Main Outcome and MeasureThe cNfL levels adjusted for age and sex across diagnoses. ResultsData were collected for 10059 individuals (mean [SD] age, 59.7 [18.8] years; 54.1% female). Thirty-five diagnoses were identified, including inflammatory diseases of the central nervous system (n=2795), dementias and predementia stages (n=4284), parkinsonian disorders (n=984), and HC (n=1332). The cNfL was elevated compared with HC in a majority of neurological conditions studied. Highest levels were observed in cognitively impaired HIV-positive individuals (iHIV), amyotrophic lateral sclerosis, frontotemporal dementia (FTD), and Huntington disease. In 33.3% of diagnoses, including HC, multiple sclerosis, Alzheimer disease (AD), and Parkinson disease (PD), cNfL was higher in men than women. The cNfL increased with age in HC and a majority of neurological conditions, although the association was strongest in HC. The cNfL overlapped in most clinically similar diagnoses except for FTD and iHIV, which segregated from other dementias, and PD, which segregated from atypical parkinsonian syndromes. Conclusions and RelevanceThese data support the use of cNfL as a biomarker of neuroaxonal damage and indicate that age-specific and sex-specific (and in some cases disease-specific) reference values may be needed. The cNfL has potential to assist the differentiation of FTD from AD and PD from atypical parkinsonian syndromes.
- Published
- 2019
31. Ultrasensitive immunoassay allows measurement of serum neurofilament heavy in multiple sclerosis
- Author
-
Marleen J. A. Koel-Simmelink, Charlotte E. Teunissen, Joep Killestein, Carsten Korth, Claire Bridel, Hugo Vrenken, Inge M.W. Verberk, Harry Twaalfhoven, Laboratory Medicine, Amsterdam Neuroscience - Neurodegeneration, Radiology and nuclear medicine, Amsterdam Neuroscience - Brain Imaging, Amsterdam Neuroscience - Neuroinfection & -inflammation, Neurology, and AII - Inflammatory diseases
- Subjects
medicine.medical_specialty ,Multiple Sclerosis ,Neurofilament ,Coefficient of variation ,Neurofilament light ,Disease duration ,Intermediate Filaments ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Neurofilament Proteins ,Internal medicine ,Linear regression ,Humans ,Medicine ,030212 general & internal medicine ,Immunoassay ,medicine.diagnostic_test ,business.industry ,Multiple sclerosis ,General Medicine ,medicine.disease ,Neurology ,Biomarker (medicine) ,Neurology (clinical) ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Background: Neurofilament heavy (NfH) is a promising biomarker for neuro-axonal damage in Multiple Sclerosis (MS). We compared the performance of high-sensitivity serum-NfH immunoassays, with as aim to investigate the value of serum-NfH as biomarker for MS. Methods: We measured serum-NfH in 76 MS patients with Simoa (one commercial, one in-house) or Luminex assays. Serum-NfH measured by the immunoassay with greatest sensitivity was related to clinical and radiological outcomes with age and sex-adjusted linear regression analysis, and to biological outcomes cerebrospinal fluid (CSF)-NfH, serum neurofilament light (NfL) and CSF-NfL with Spearman's correlation analysis. Results: With the commercial Simoa assay, we obtained 100% serum-NfH detectability (in-house Simoa: 70%, Luminex: 61%), with lowest coefficient of variation (CV) between duplicates of 11%CV (in-house Simoa: 22%CV, Luminex: 30%CV). Serum-NfH quantified with the commercial Simoa assay was associated with disease duration (standardized beta (sβ) = 0.28, p = 0.034), T2 lesion volume (sβ = 0.23, p = 0.041), and tended to associate with black hole count (sβ = 0.21, p = 0.084) but not with Expanded Disease Disability Score (EDSS) or normalized brain volume (all: p>0.10). Furthermore, serum-NfH showed correlations with CSF-NfH (rho = 0.27, p = 0.018) and serum-NfL (rho=0.44, p < 0.001), but not with CSF-NfL. Conclusions: Serum-NfH can be quantified with high-sensitivity technology. Cross-sectionally, we observed some weak correlations of serum-NfH with MS disease burden parameters, suggesting there might be some utility for serum-NfH as biomarker for MS disease burden.
- Published
- 2021
32. P1‐247: CEREBROSPINAL FLUID NEUROFILAMENT LIGHT PROTEIN AS A DIFFERENTIAL DIAGNOSIS BIOMARKER IN NEUROLOGICAL DISEASES: A SYSTEMATIC REVIEW AND METAANALYSIS
- Author
-
Pieter Jelle Visser, Henrik Zettenberg, Charlotte E. Teunissen, Wessel N. van Wieringen, Claire Bridel, and Betty M. Tijms
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Epidemiology ,business.industry ,Health Policy ,Neurofilament light ,03 medical and health sciences ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,Cerebrospinal fluid ,Developmental Neuroscience ,Biomarker (medicine) ,Medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,Differential diagnosis ,business ,030217 neurology & neurosurgery - Published
- 2018
33. P2‐228: PRE‐ANALYTICAL STABILITY OF NOVEL CEREBROSPINAL FLUID BIOMARKERS FOR DEMENTIA
- Author
-
Vera M. Mendes, Wiesje M. Flier, Yannick Vermeiren, Naomi De Roeck, Peter Paul De Deyn, Javier Sáez-Valero, Charlotte E. Teunissen, Eduard A. Struys, Ulf Andreasson, Erwin Janssen, Eugeen Vanmechelen, María-Salud García-Ayllón, Kees Wj. Uffelen, Bruno Manadas, Eline A.J. Willemse, Claire Bridel, Sebastiaan Engelborghs, and Inmaculada Lopes Font
- Subjects
Oncology ,medicine.medical_specialty ,Epidemiology ,Pre analytical ,business.industry ,Health Policy ,medicine.disease ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Cerebrospinal fluid ,Developmental Neuroscience ,Internal medicine ,medicine ,Dementia ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2018
34. No Plasmatic Proteomic Signature at Clinical Disease Onset Associated With 11 Year Clinical, Cognitive and MRI Outcomes in Relapsing-Remitting Multiple Sclerosis Patients
- Author
-
Charlotte E. Teunissen, Joep Killestein, Cyra E. Leurs, Anand J. C. Eijlers, Claire Bridel, Menno M. Schoonheim, Wessel N. van Wieringen, Marleen J.A. Koel-Simmelink, Mathematics, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, Amsterdam Neuroscience - Complex Trait Genetics, Clinical chemistry, Anatomy and neurosciences, Amsterdam Neuroscience - Neuroinfection & -inflammation, NCA - Neuroinflamation, Epidemiology and Data Science, NCA - Brain mechanisms in health and disease, Other Research, Neurology, and AII - Inflammatory diseases
- Subjects
0301 basic medicine ,Oncology ,Proteomics ,cognition ,medicine.medical_specialty ,multiple sclerosis ,lcsh:RC321-571 ,Multiple sclerosis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Cerebrospinal fluid ,Cognition ,proteomics ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Molecular Biology ,business.industry ,Clinical course ,Brief Research Report ,medicine.disease ,Clinical disease ,Prognosis ,030104 developmental biology ,Relapsing remitting ,Proteome ,prognosis ,business ,030217 neurology & neurosurgery ,Neuroscience ,MRI - Abstract
Background: The clinical course of relapsing-remitting multiple sclerosis (RRMS) is highly heterogeneous and prognostic biomarkers at time of diagnosis are lacking. Objective: We investigated the predictive value of the plasma proteome at time of diagnosis in RRMS patients. Methods: The plasma proteome was interrogated using a novel aptamer-based proteomics platform, which allows to measure the levels of a predefined set of 1310 proteins. Results: In 67 clinically and radiologically well characterized RRMS patients, we found no association between the plasma proteome at diagnosis and clinical, cognitive or MRI outcomes after 11 years. Conclusions: Proteomics studies on cerebrospinal fluid may be better suited to identify prognostic biomarkers in early RRMS.
- Published
- 2018
35. Regulator of oligodendrocyte maturation, miR-219, a potential biomarker for MS
- Author
-
Joep Killestein, Claire Bridel, Lawrence Steinman, Rogier Q. Hintzen, Ilona B. Bruinsma, Marcel M. Verbeek, Tessel F. Runia, Marie van Dijk, Brigit A. de Jong, Timothy van de Lisdonk, Charlotte E. Teunissen, Sanne Q. Haverkort, Neurology, Immunology, Amsterdam Reproduction & Development (AR&D), ACS - Atherosclerosis & ischemic syndromes, Laboratory Medicine, Amsterdam Neuroscience - Neuroinfection & -inflammation, and APH - Quality of Care
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,miR-219 ,Immunology ,Central nervous system ,lcsh:RC346-429 ,Multiple sclerosis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,miR-150 ,0302 clinical medicine ,Degenerative disease ,Cerebrospinal fluid ,Precursor cell ,Humans ,Medicine ,Remyelination ,lcsh:Neurology. Diseases of the nervous system ,miRNA ,business.industry ,Research ,General Neuroscience ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,Oligodendrocyte ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Biomarker (medicine) ,Female ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Contains fulltext : 182383.pdf (Publisher’s version ) (Open Access) BACKGROUND: Multiple sclerosis (MS) is a demyelinating and degenerative disease of the central nervous system. Normally, demyelination is followed by remyelination, which requires repopulation of a demyelinated area by oligodendrocyte precursor cells. Although large numbers of precursor cells are present in MS lesions, remyelination often fails, in part by the inability of precursor cells to differentiate into mature myelin-forming cells. In mouse and rat, miR-219 is required for this differentiation. Previously, we identified decreased miR-219 expression in tissue of MS patients compared to controls. Cell-free miRNAs have been detected in many different body fluids including cerebrospinal fluid (CSF) and may reflect disease processes going on in the central nervous system. This prompted us to investigate the biomarker performance of CSF miR-219 for MS diagnosis. METHODS: Quantitative PCR was performed measuring miR-219 levels in CSF of MS patients and controls in three independent cohorts. RESULTS: All three cohorts of MS patients and controls revealed that absence of miR-219 detection in CSF is consistently associated with MS. CONCLUSIONS: We have been able to identify and validate absence of miR-219 detection in CSF of MS patients compared to controls, suggesting that it may emerge as a candidate biomarker for MS diagnosis.
- Published
- 2017
36. [P1–276]: BRI2 DEMENTIA PATHWAY IN CSF, IMPROVING DETECTION THROUGH ULTRASENSITIVE SIMOA TECHNOLOGY
- Author
-
Philip Scheltens, Claire Bridel, Harry Twaalfhoven, Marta Del Campo, Charlotte E. Teunissen, and Lynn Boonkamp
- Subjects
Oncology ,medicine.medical_specialty ,Epidemiology ,business.industry ,Health Policy ,medicine.disease ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Internal medicine ,medicine ,Dementia ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2017
37. Glutaminyl cyclase activity correlates with levels of Aβ peptides and mediators of angiogenesis in cerebrospinal fluid of Alzheimer’s disease patients
- Author
-
Sisi Durieux, Antje Meyer, Philip Scheltens, Claire Bridel, Inge Lues, Marleen J. A. Koel-Simmelink, Matthias Orth, Charlotte E. Teunissen, Torsten Hoffmann, Clinical chemistry, Neurology, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
0301 basic medicine ,Apolipoprotein E ,Male ,Neurology ,Angiogenesis ,Statistics as Topic ,lcsh:RC346-429 ,0302 clinical medicine ,Cerebrospinal fluid ,Angiogenic Proteins ,Aged, 80 and over ,biology ,Angiogenesis Modulating Agents ,Middle Aged ,Glutaminyl cyclase ,Aminoacyltransferases ,Angiopoietin receptor ,Disease Progression ,Biomarker (medicine) ,Female ,Alzheimer’s disease ,medicine.medical_specialty ,Amyloid beta ,Cognitive Neuroscience ,Sensitivity and Specificity ,lcsh:RC321-571 ,03 medical and health sciences ,Alzheimer Disease ,Internal medicine ,medicine ,3pE-Aβ42 ,Humans ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,lcsh:Neurology. Diseases of the nervous system ,Aged ,Amyloid beta-Peptides ,business.industry ,Research ,Reproducibility of Results ,Enzyme Activation ,030104 developmental biology ,Endocrinology ,Pharmacodynamics ,biology.protein ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Background Pyroglutamylation of truncated Aβ peptides, which is catalysed by enzyme glutaminyl cyclase (QC), generates pE-Aβ species with enhanced aggregation propensities and resistance to most amino-peptidases and endo-peptidases. pE-Aβ species have been identified as major constituents of Aβ plaques and reduction of pE-Aβ species is associated with improvement of cognitive tasks in animal models of Alzheimer’s disease (AD). Pharmacological inhibition of QC has thus emerged as a promising therapeutic approach for AD. Here, we question whether cerebrospinal fluid (CSF) QC enzymatic activity differs between AD patients and controls and whether inflammatory or angiogenesis mediators, some of which are potential QC substrates, and/or Aβ peptides may serve as pharmacodynamic read-outs for QC inhibition. Methods QC activity, Aβ peptides and inflammatory or angiogenesis mediators were measured in CSF of a clinically well-characterized cohort of 20 mild AD patients, 20 moderate AD patients and 20 subjective memory complaints (SMC) controls. Correlation of these parameters with core diagnostic CSF AD biomarkers (Aβ42, tau and p-tau) and clinical features was evaluated. Results QC activity shows a tendency to decrease with AD progression (p = 0.129). The addition of QC activity to biomarkers tau and p-tau significantly increases diagnostic power (ROC-AUCTAU = 0.878, ROC-AUCTAU&QC = 0.939 and ROC-AUCpTAU = 0.820, ROC-AUCpTAU&QC = 0.948). In AD and controls, QC activity correlates with Aβ38 (r = 0.83, p 0.5, p 0.35, p =
- Published
- 2017
38. Correction: Aerosols transmit prions to immunocompetent and immunodeficient mice
- Author
-
Mathias Heikenwalder, Elizabeta Zirdum, Adriano Aguzzi, Thomas J. Fuchs, Britta Klevenz, Benjamin Petsch, Petra Schwarz, Ilan Margalith, Kirsten D. Mertz, Claire Bridel, Johannes Haybaeck, Lothar Stitz, and University of Zurich
- Subjects
0301 basic medicine ,Male ,lcsh:Immunologic diseases. Allergy ,Prions ,Immunology ,Longevity ,10208 Institute of Neuropathology ,610 Medicine & health ,Mice, Inbred Strains ,Mice, Transgenic ,Mice, SCID ,Biology ,Microbiology ,03 medical and health sciences ,Immunocompromised Host ,Mice ,Species Specificity ,Prion infectivity ,Virology ,Genetics ,Animals ,Molecular Biology ,lcsh:QH301-705.5 ,Aerosols ,Mice, Knockout ,Neurons ,Inhalation Exposure ,Correction ,Brain ,030104 developmental biology ,Animals, Newborn ,lcsh:Biology (General) ,570 Life sciences ,biology ,Parasitology ,Female ,lcsh:RC581-607 ,Immunocompetence ,Scrapie - Abstract
The authors would like to correct Figs Figs3,3, ,55 and and6.6. In Fig 3, an error was introduced during the preparation of the figure for publication. Images from the brain of a mouse presented in Fig 4A were inserted into Fig 3A. The corrected version of Fig 3, containing pictures of a correct and representative JH-/- mouse that had been exposed to prion infectivity containing aerosols, can be seen here. The authors also wish to clarify that the original blots for Figs Figs5E5E and and6D6D contained redundant lanes which they had removed from the images while preparing the figures. The authors now provide corrected versions of Figs Figs5E5E and and6D6D with appropriate marks showing the removal of the redundant lane. The uncropped original blots for Figs Figs5E5E and and6D6D are shown as supporting information in S1 File (for Fig 5E) and S2 File (for Fig 6D). Fig 3 Prion transmission through aerosols in immunocompromised mice. Fig 5 Prion transmission by intranasal instillation. Fig 6 Intranasal prion transmission in immnunodeficient mice. The corrected versions of Figs Figs3,3, ,55 and and66 and the original uncropped blots for Figs Figs5E5E and and6D6D can be viewed below. The authors confirm that these changes do not alter their findings.
- Published
- 2016
39. Multifocal motor neuropathy with high titers of anti-MAG antibodies
- Author
-
Claire Bridel, Judit Horvath, Andreas J. Steck, André Truffert, Mary Kurian, and Patrice H. Lalive
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,General Neuroscience ,Mismatch negativity ,Context (language use) ,Overlap syndrome ,Tetraparesis ,medicine.disease ,medicine.anatomical_structure ,Skin biopsy ,medicine ,Terminal nerve ,Neurology (clinical) ,Perineurium ,business ,Multifocal motor neuropathy - Abstract
Multifocal motor neuropathy (MMN) and anti-myelin-associated glycoprotein (anti-MAG)-associated neuropathy are clinically and electrophysiologically distinct entities. We describe a patient with characteristic features of both neuropathies, raising the possibility of an overlap syndrome. A 49-year-old patient reported a history of slowly progressive predominantly distal tetraparesis, with mild sensory deficits. Nerve conduction studies demonstrated persistent motor conduction blocks outside compression sites, typical of MMN. Laboratory findings revealed persistently high titers of anti-MAG immunoglobulin Mλ (IgMλ) paraprotein in the context of a monoclonal gammapathy of unknown significance. Skin biopsy of distal lower limb revealed IgM positive terminal nerve perineurium deposits. This case suggests that the distinction between subtypes of chronic inflammatory neuropathies may not be as clear as initially thought, and that the pattern of pathogenicity of anti-MAG antibodies may vary.
- Published
- 2014
40. Teaching NeuroImages: Drug-induced parkinsonism with asymmetrical putaminal DaT binding
- Author
-
Valentina Garibotto and Claire Bridel
- Subjects
medicine.medical_specialty ,ddc:616.0757 ,Parkinsonian Disorders ,Internal medicine ,Basal ganglia ,medicine ,Humans ,Infarction, Middle Cerebral Artery/complications ,Spasticity ,Stroke ,Tomography, Emission-Computed, Single-Photon ,Dopamine Plasma Membrane Transport Proteins ,Risperidone ,Parkinsonism ,Putamen ,Putamen/metabolism ,Sequela ,Infarction, Middle Cerebral Artery ,Middle Aged ,medicine.disease ,Gait ,Risperidone/adverse effects ,Cardiology ,Etiology ,Neurology (clinical) ,medicine.symptom ,Psychology ,Dopamine Plasma Membrane Transport Proteins/metabolism ,Parkinsonian Disorders/chemically induced/diagnostic imaging/metabolism ,medicine.drug - Abstract
A 64-year-old patient developed shuffling gait and postural instability over 8 months, in parallel to risperidone intake. Examination showed symmetric parkinsonian syndrome and mild left hemiparesis with spasticity, related to right middle cerebral artery stroke 10 years before. Within 1 month of risperdone withdrawal, parkinsonism disappeared, confirming drug-induced etiology. While degenerative parkinsonism is associated with reduction in striatal dopamine transporters binding in absence of structural lesions to basal ganglia, binding in drug-induced parkinsonism is normal.1 In our case, SPECT anomaly was incidental, related to the ischemic sequela (figure).2 This case highlights the importance of brain structural integrity for SPECT interpretation.
- Published
- 2015
41. A phase IIa randomized clinical study testing GNbAC1, a humanized monoclonal antibody against the envelope protein of multiple sclerosis associated endogenous retrovirus in multiple sclerosis patients - a twelve month follow-up
- Author
-
Claire Bridel, Alois B. Lang, Hervé Porchet, Maria Rasenack, François Curtin, Raphaël Faucard, Ludwig Kappos, Jules Alexandre Desmeules, Hervé Perron, Patrice H. Lalive, Alain Matthey, Tobias Derfuss, Myriam Schluep, Claudia Guebelin, Hans-Peter Hartung, and Renaud Du Pasquier
- Subjects
Male ,Multiple Sclerosis ,medicine.drug_class ,Immunology ,Humanized/pharmacology/therapeutic use ,Endogenous retrovirus ,Endogeny ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Antibodies ,Cohort Studies ,Pharmacokinetics ,Viral Envelope Proteins ,Monoclonal ,Immunology and Allergy ,Medicine ,Humans ,Aged ,ddc:615 ,ddc:617 ,biology ,business.industry ,Multiple sclerosis ,Endogenous Retroviruses ,Middle Aged ,medicine.disease ,Endogenous Retroviruses/drug effects/immunology ,Clinical trial ,Neurology ,Pharmacodynamics ,biology.protein ,Female ,Neurology (clinical) ,Antibody ,Viral Envelope Proteins/antagonists & inhibitors/immunology ,business ,Multiple Sclerosis/drug therapy/immunology ,Follow-Up Studies - Abstract
GNbAC1 is a humanized monoclonal antibody targeting MSRV-Env, an endogenous retroviral protein, which is expressed in multiple sclerosis (MS) lesions, is pro-inflammatory and inhibits oligodendrocyte precursor cell differentiation. This paper describes the open-label extension up to 12months of a trial testing GNbAC1 in 10 MS patients at 2 and 6mg/kg. The primary objective was to assess GNbAC1 safety, and other objectives were pharmacokinetic and pharmacodynamic assessments. During the extended study, no safety issues occurred in the 8 remaining patients. No anti-GNbAC1 antibodies were detected. GNbAC1 appears well tolerated.
- Published
- 2015
42. Cerebrospinal fluid angiotensin-converting enzyme for diagnosis of neurosarcoidosis
- Author
-
Nicolas Vuilleumier, Patrice H. Lalive, Claire Bridel, and Delphine S. Courvoisier
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Sarcoidosis ,Immunology ,Central Nervous System Diseases/cerebrospinal fluid/diagnosis ,Peptidyl-Dipeptidase A ,Gastroenterology ,Cerebrospinal fluid ,Central Nervous System Diseases ,Internal medicine ,Immunology and Allergy ,Medicine ,Humans ,ddc:576 ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,Receiver operating characteristic ,business.industry ,Lumbar puncture ,Multiple sclerosis ,Area under the curve ,Neurosarcoidosis ,Biological Markers/cerebrospinal fluid ,Middle Aged ,medicine.disease ,Neurology ,Female ,Sarcoidosis/cerebrospinal fluid/diagnosis ,Neurology (clinical) ,business ,Vasculitis ,Biomarkers ,Peptidyl-Dipeptidase A/cerebrospinal fluid - Abstract
Background Neurosarcoidosis (NS) is a rare condition that may mimic central nervous system (CNS) infection, neoplasia and other inflammatory disorders of the CNS such as multiple sclerosis, encephalitis and vasculitis. Diagnosis is challenging in cases with minimal or absent systemic involvement. Cerebrospinal fluid (CSF) angiotensin-converting enzyme (c-ACE) has been claimed as a valuable diagnostic tool for NS. However, there is little data evaluating its performance in routine clinical practice. Findings We performed a monocentric, retrospective, chart-based study including all patients investigated with a lumbar puncture and c-ACE dosage for suspected NS between 01/01/2006 and 31/12/2012 at the Geneva University Hospital. Receiver-operating characteristic (ROC) curve and area under the curve (AUC) were performed to calculate the optimal cut-off value of c-ACE and to determine the discriminative ability of c-ACE. Of the 440 patients included in the study, 9 were diagnosed with NS on the basis of tissue biopsy. Mean c-ACE was not significantly different between NS and non-NS patients. With a cut-off value of 2 (0–2 vs ≥ 3), sensitivity and specificity of c-ACE were 66.7% and 67.3%, respectively. Conclusions In our clinical setting, the sensitivity and specificity of c-ACE for NS diagnosis were relatively poor and of little clinical utility.
- Published
- 2015
43. Update on multiple sclerosis treatments
- Author
-
Patrice H. Lalive and Claire Bridel
- Subjects
medicine.medical_specialty ,Daclizumab ,Toluidines ,Dimethyl Fumarate ,Hydroxybutyrates ,Disease ,Interferon-beta/therapeutic use ,Immunoglobulin G/therapeutic use ,Antibodies, Monoclonal, Humanized ,chemistry.chemical_compound ,Natalizumab ,Multiple Sclerosis, Relapsing-Remitting ,Fumarates ,Sphingosine ,Crotonates/therapeutic use ,Teriflunomide ,Nitriles ,Medicine ,Humans ,Glatiramer acetate ,Propylene Glycols/therapeutic use ,Intensive care medicine ,Alemtuzumab ,business.industry ,Fingolimod Hydrochloride ,Fumarates/therapeutic use ,Multiple sclerosis ,General Medicine ,Interferon-beta ,Multiple Sclerosis, Relapsing-Remitting/drug therapy ,medicine.disease ,Fingolimod ,ddc:616.8 ,Antibodies, Monoclonal, Humanized/therapeutic use ,chemistry ,Toluidines/therapeutic use ,Propylene Glycols ,Crotonates ,Immunoglobulin G ,Immunosuppressive Agents/administration & dosage/adverse effects/therapeutic use ,Sphingosine/analogs & derivatives/therapeutic use ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
Relapsing-remitting multiple sclerosis (RRMS) management has dramatically changed over the past decade. New drugs have arrived on the market, allowing for more individualised treatment selection. However, this diversity has increased the complexity of RRMS patient follow-up. In this review, we provide summarised information about treatment efficacy, potential side-effects, follow-up recommendations, vaccinations, and pregnancy safety issues for all currently available disease modifying therapies and those awaiting approval.
- Published
- 2014
44. A phase IIa randomised clinical study of GNbAC1, a humanised monoclonal antibody against the envelope protein of multiple sclerosis-associated endogenous retrovirus in multiple sclerosis patients
- Author
-
Ludwig Kappos, Maria Rasenack, François Curtin, Patrice H. Lalive, Hans-Peter Hartung, Tobias Derfuss, Alois B. Lang, Claire Bridel, Alain Matthey, Claudia Guebelin, Renaud Du Pasquier, Hervé Porchet, Raphaël Faucard, Jules Alexandre Desmeules, Myriam Schluep, and Hervé Perron
- Subjects
Adult ,Male ,Multiple Sclerosis ,medicine.drug_class ,Placebo ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Polymerase Chain Reaction ,Retrovirus ,Pharmacokinetics ,Double-Blind Method ,Medicine ,Humans ,biology ,ddc:617 ,business.industry ,Immunogenicity ,Multiple sclerosis ,Endogenous Retroviruses ,Brain ,Gene Products, env ,Middle Aged ,biology.organism_classification ,medicine.disease ,Magnetic Resonance Imaging ,Neurology ,Pharmacodynamics ,Immunology ,biology.protein ,RNA, Viral ,Female ,Neurology (clinical) ,Antibody ,business - Abstract
Background: GNbAC1 is an immunoglobulin (IgG4) humanised monoclonal antibody against multiple sclerosis-associated retrovirus (MSRV)-Env, a protein of endogenous retroviral origin, expressed in multiple sclerosis (MS) lesions, which is pro-inflammatory and inhibits oligodendrocyte precursor cell differentiation. Objective: This is a randomised, double-blind placebo-controlled dose-escalation study followed by a six-month open-label phase to test GNbAC1 in MS patients. The primary objective was to assess GNbAC1 safety in MS patients, and the other objectives were pharmacokinetic and pharmacodynamic assessments. Methods: Ten MS patients were randomised into two cohorts to receive a single intravenous infusion of GNbAC1/placebo at doses of 2 or 6 mg/kg. Then all patients received five infusions of GNbAC1 at 2 or 6 mg/kg at four-week intervals in an open-label setting. Safety, brain magnetic resonance imaging (MRI), pharmacokinetics, immunogenicity, cytokines and MSRV RNA expression were studied. Results: All patients completed the study. GNbAC1 was well tolerated in all patients. GNbAC1 pharmacokinetics is dose-linear with mean elimination half-life of 27–37 d. Anti-GNbAC1 antibodies were not detected. Cytokine analysis did not indicate an adverse effect. MSRV-transcripts showed a decline after the start of treatment. Nine patients had stable brain lesions at MRI. Conclusion: The safety, pharmacokinetic profile, and pharmacodynamic responses to GNbAC1 are favourable in MS patients over a six-month treatment period.
- Published
- 2014
45. Minimal supportive treatment in natalizumab-related PML in a MS patient
- Author
-
Laurent Kaiser, Frederik Barkhof, Ruxandra Iancu Ferfoglia, Patrice H. Lalive, Sven Haller, R. Du Pasquier, Claire Bridel, Radiology and nuclear medicine, and NCA - Neuroinflamation
- Subjects
Palliative care ,JC virus ,medicine.disease_cause ,Antibodies, Monoclonal, Humanized/adverse effects/therapeutic use ,ddc:616.0757 ,Multiple Sclerosis, Relapsing-Remitting/diagnosis/drug therapy ,Disability Evaluation ,Neurologic Examination/drug effects ,Natalizumab ,Immune system ,Immune reconstitution inflammatory syndrome ,parasitic diseases ,medicine ,Humans ,Leukoencephalopathy, Progressive Multifocal/diagnosis/drug therapy ,ddc:616 ,biology ,business.industry ,Progressive multifocal leukoencephalopathy ,Palliative Care ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,ddc:616.8 ,Brain/drug effects/pathology ,Psychiatry and Mental health ,Diffusion Magnetic Resonance Imaging ,Monoclonal ,Immunology ,biology.protein ,Disease Progression ,Surgery ,Female ,Neurology (clinical) ,Antibody ,Immune Reconstitution Inflammatory Syndrome/chemically induced/diagnosis ,business ,medicine.drug ,Follow-Up Studies - Abstract
Natalizumab (NTZ) is a recombinant humanised immunoglobulin G4 monoclonal antibody that selectively inhibits adhesion of α4-β1 receptor on the surface of lymphocytes and hinders circulating cells from leaving the vascular compartment of the brain.1 NTZ is associated with a risk of developing progressive multifocal leukoencephalopathy (PML), estimated at approximately 1/200 in JCV-positive patients after 24 months treatment duration.1 The overall survival rate is 71% in PML secondary to NTZ treatment, which is substantially better than the often-fatal course of PML in other immunosuppressed patients.2 This reflects the fact that immune reconstitution is more easily obtained in NTZ patient by treatment cessation.3 Immune reconstitution inflammatory syndrome (IRIS), characterised by contrast-enhancing MRI lesions with clinical deterioration, is observed in almost all PML patients after NTZ cessation.2 Plasma exchange (PLEX) is usually initiated after NTZ cessation to remove remaining circulating NTZ levels, which favours the immune reconstitution, whereas intravenous corticosteroids (CS) are given to suppress IRIS development.1 Nevertheless, the increased peripheral functional T-cell expansion following PLEX can be seen as an accelerator for IRIS, and the negative impact of CS on JCV-specific CD8 T cell response is also at risk of inhibiting the physiological immune response against the JC virus (JCV). ### Report of a case A patient was diagnosed with relapsing-remitting MS at the age of 46 years. Her JCV IgG serum status was positive 6 months after NTZ treatment initiation. After 22 infusions, she presented …
- Published
- 2015
46. Aerosols Transmit Prions to Immunocompetent and Immunodeficient Mice
- Author
-
Kirsten D. Mertz, Claire Bridel, Elizabeta Zirdum, Thomas J. Fuchs, Petra Schwarz, Johannes Haybaeck, Mathias Heikenwalder, Benjamin Petsch, Lothar Stitz, Ilan Margalith, Adriano Aguzzi, Britta Klevenz, University of Zurich, and Aguzzi, A
- Subjects
Genetically modified mouse ,QH301-705.5 ,Transgene ,animal diseases ,Immunology ,10208 Institute of Neuropathology ,2405 Parasitology ,Spleen ,Scrapie ,610 Medicine & health ,Biology ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Inbred strain ,1311 Genetics ,Virology ,Genetics ,medicine ,1312 Molecular Biology ,Biology (General) ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,2403 Immunology ,Follicular dendritic cells ,2404 Microbiology ,RC581-607 ,3. Good health ,nervous system diseases ,medicine.anatomical_structure ,Lymphatic system ,Pathology/Neuropathology ,2406 Virology ,570 Life sciences ,biology ,Parasitology ,Immunocompetence ,Immunologic diseases. Allergy ,030217 neurology & neurosurgery ,Research Article ,Neuroscience - Abstract
Prions, the agents causing transmissible spongiform encephalopathies, colonize the brain of hosts after oral, parenteral, intralingual, or even transdermal uptake. However, prions are not generally considered to be airborne. Here we report that inbred and crossbred wild-type mice, as well as tga20 transgenic mice overexpressing PrPC, efficiently develop scrapie upon exposure to aerosolized prions. NSE-PrP transgenic mice, which express PrPC selectively in neurons, were also susceptible to airborne prions. Aerogenic infection occurred also in mice lacking B- and T-lymphocytes, NK-cells, follicular dendritic cells or complement components. Brains of diseased mice contained PrPSc and transmitted scrapie when inoculated into further mice. We conclude that aerogenic exposure to prions is very efficacious and can lead to direct invasion of neural pathways without an obligatory replicative phase in lymphoid organs. This previously unappreciated risk for airborne prion transmission may warrant re-thinking on prion biosafety guidelines in research and diagnostic laboratories., Author Summary Prions, which are the cause of fatal neurodegenerative disorders termed transmissible spongiform encephalopathies (TSEs), can be experimentally or naturally transmitted via prion-contaminated food, blood, milk, saliva, feces and urine. Here we demonstrate that prions can be transmitted through aerosols in mice. This also occurs in the absence of immune cells as demonstrated by experiments with mice lacking B-, T-, follicular dendritic cells (FDCs), lymphotoxin signaling or with complement-deficient mice. Therefore, a functionally intact immune system is not strictly needed for aerogenic prion infection. These results suggest that current biosafety guidelines applied in diagnostic and scientific laboratories ought to include prion aerosols as a potential vector for prion infection.
- Published
- 2011
- Full Text
- View/download PDF
47. Follicular dendritic cells control engulfment of apoptotic bodies by secreting Mfge8
- Author
-
Anita Schildknecht, Jan Kranich, Marie Kosco-Vilbois, Gino Miele, Adriano Aguzzi, Christoph Huber, Magdalini Polymenidou, Ernst Heinen, Rolf M. Zinkernagel, Claire Bridel, Nike Julia Krautler, University of Zurich, and Aguzzi, A
- Subjects
Immunology ,10208 Institute of Neuropathology ,Spleen ,Apoptosis ,Mice, Inbred Strains ,610 Medicine & health ,Biology ,medicine.disease_cause ,Antibodies ,Autoimmunity ,Mice ,Antigen ,Epidermal growth factor ,medicine ,In Situ Nick-End Labeling ,Immunology and Allergy ,Animals ,Crosses, Genetic ,In Situ Hybridization ,Bone Marrow Transplantation ,DNA Primers ,Mice, Knockout ,B-Lymphocytes ,2403 Immunology ,Follicular dendritic cells ,Macrophages ,Brief Definitive Report ,Germinal center ,Dendritic Cells ,Milk Proteins ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Antigens, Surface ,biology.protein ,2723 Immunology and Allergy ,Leukocyte Common Antigens ,RNA ,Brief Definitive Reports ,570 Life sciences ,biology ,Receptors, Complement 3d ,Antibody ,MFGE8 ,Dendritic Cells, Follicular - Abstract
The secreted phosphatidylserine-binding protein milk fat globule epidermal growth factor 8 (Mfge8) mediates engulfment of apoptotic germinal center B cells by tingible-body macrophages (TBMphis). Impairment of this process can contribute to autoimmunity. We show that Mfge8 is identical to the mouse follicular dendritic cell (FDC) marker FDC-M1. In bone-marrow chimeras between wild-type and Mfge8(-/-) mice, all splenic Mfge8 was derived from FDCs rather than TBMphis. However, Mfge8(-/-) TBMphis acquired and displayed Mfge8 only when embedded in Mfge8(+/+) stroma, or when situated in lymph nodes draining exogenous recombinant Mfge8. These findings indicate a licensing role for FDCs in TBMphi-mediated removal of excess B cells. Lymphotoxin-deficient mice lacked FDCs and splenic Mfge8, and suffer from autoimmunity similar to Mfge8(-/-) mice. Hence, FDCs facilitate TBMphi-mediated corpse removal, and their malfunction may be involved in autoimmunity.
- Published
- 2008
48. Hematologic modifications in natalizumab-treated multiple sclerosis patients
- Author
-
Kaveh Samii, Patrice H. Lalive, Claire Bridel, and Yan Beauverd
- Subjects
medicine.medical_specialty ,Lymphocyte ,ddc:616.07 ,Hematocrit ,Bioinformatics ,Gastroenterology ,Article ,Blood cell ,Natalizumab ,immune system diseases ,White blood cell ,Internal medicine ,medicine ,Acute leukemia ,Hematology ,medicine.diagnostic_test ,business.industry ,Multiple sclerosis ,medicine.disease ,nervous system diseases ,medicine.anatomical_structure ,Neurology ,Neurology (clinical) ,business ,medicine.drug - Abstract
Objective: To monitor the hematologic modifications in the peripheral blood of patients with relapsing-remitting multiple sclerosis treated with natalizumab. Methods: The cohort included 44 patients with relapsing-remitting multiple sclerosis treated monthly with natalizumab for 18 months. Peripheral blood was collected before treatment initiation and on a monthly basis during the treatment course. Complete blood cell count was performed using automated hematology systems. Blood smears were prepared and analyzed when abnormal values were detected. Results: Mean total white blood cell, lymphocyte, and eosinophil counts were significantly higher 1 month after treatment initiation and remained stable during the 18 months of follow-up. Monocyte counts increased progressively during the 18-month treatment with natalizumab. Erythroblasts and neutrophil precursors were absent before treatment initiation but were present in 16% and 6.8% of patients, respectively, 1 month after the first natalizumab infusion. The proportion of patients with erythroblasts and neutrophil precursors remained stable throughout the 18-month follow-up period. On an individual patient basis, a fluctuating level of erythroblasts and neutrophil precursors was observed. No difference in mean erythrocyte, hemoglobin, hematocrit, thrombocyte, and neutrophil levels was observed before and after 18 months of natalizumab treatment. No cases of myelodysplastic syndrome or acute leukemia were observed. Conclusion: Chronic treatment with natalizumab is associated with significant modifications in complete blood cell count, including emergence of hematopoietic precursors that are not present in peripheral blood under normal conditions. None of these modifications were associated with malignancy.
- Published
- 2015
49. Neurodegeneration and unfolded-protein response in mice expressing a membrane-tethered flexible tail of PrP
- Author
-
Tracy O'Connor, Asvin K. K. Lakkaraju, Donal McHugh, Claire Bridel, Pawel Pelczar, Thomas Rülicke, Paolo Dametto, Arlind Adili, Lukas Villiger, Uli S. Herrmann, Adriano Aguzzi, and University of Zurich
- Subjects
Genetically modified mouse ,Prions ,Immunoprecipitation ,Allosteric regulation ,10208 Institute of Neuropathology ,lcsh:Medicine ,Mice, Transgenic ,610 Medicine & health ,1100 General Agricultural and Biological Sciences ,Biology ,Prion Diseases ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,1300 General Biochemistry, Genetics and Molecular Biology ,Cerebellum ,medicine ,Animals ,PrPC Proteins ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,1000 Multidisciplinary ,Multidisciplinary ,Endoplasmic reticulum ,lcsh:R ,Neurodegeneration ,Endoplasmic Reticulum Stress ,medicine.disease ,Molecular biology ,Unfolded Protein Response ,Unfolded protein response ,Phosphorylation ,570 Life sciences ,biology ,lcsh:Q ,030217 neurology & neurosurgery ,Research Article - Abstract
The cellular prion protein (PrPC) consists of a flexible N-terminal tail (FT, aa 23-128) hinged to a membrane-anchored globular domain (GD, aa 129-231). Ligation of the GD with antibodies induces rapid neurodegeneration, which is prevented by deletion or functional inactivation of the FT. Therefore, the FT is an allosteric effector of neurotoxicity. To explore its mechanism of action, we generated transgenic mice expressing the FT fused to a GPI anchor, but lacking the GD (PrPΔ141-225, or "FTgpi"). Here we report that FTgpi mice develop a progressive, inexorably lethal neurodegeneration morphologically and biochemically similar to that triggered by anti-GD antibodies. FTgpi was mostly retained in the endoplasmic reticulum, where it triggered a conspicuous unfolded protein response specifically activating the PERK pathway leading to phosphorylation of eIF2α and upregulation of CHOP ultimately leading to neurodegeration similar to what was observed in prion infection.
- Published
- 2015
- Full Text
- View/download PDF
50. Unexpected Tolerance of α-Cleavage of the Prion Protein to Sequence Variations
- Author
-
Anna Maria Calella, José B. Oliveira-Martins, Sei-ichi Yusa, Adriano Aguzzi, Frank Baumann, Claire Bridel, Paolo Dametto, and University of Zurich
- Subjects
Glycosylation ,PrPSc Proteins ,animal diseases ,Mutant ,lcsh:Medicine ,Mice ,0302 clinical medicine ,lcsh:Science ,Peptide sequence ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,medicine.diagnostic_test ,Neurological Disorders/Prion Diseases ,Neuroscience/Neurobiology of Disease and Regeneration ,Hydrophobic and Hydrophilic Interactions ,Research Article ,Prions ,Immunoprecipitation ,Proteolysis ,Blotting, Western ,Molecular Sequence Data ,10208 Institute of Neuropathology ,610 Medicine & health ,1100 General Agricultural and Biological Sciences ,Biology ,Cleavage (embryo) ,Prion Proteins ,Cell Line ,03 medical and health sciences ,1300 General Biochemistry, Genetics and Molecular Biology ,Infectious Diseases/Prion Diseases ,medicine ,Animals ,Humans ,PrPC Proteins ,Amino Acid Sequence ,Binding site ,030304 developmental biology ,1000 Multidisciplinary ,Binding Sites ,Sequence Homology, Amino Acid ,Point mutation ,lcsh:R ,Molecular biology ,nervous system diseases ,Mutation ,NIH 3T3 Cells ,570 Life sciences ,biology ,lcsh:Q ,030217 neurology & neurosurgery ,HeLa Cells ,Peptide Hydrolases - Abstract
The cellular form of the prion protein, PrP(C), undergoes extensive proteolysis at the alpha site (109K [see text]H110). Expression of non-cleavable PrP(C) mutants in transgenic mice correlates with neurotoxicity, suggesting that alpha-cleavage is important for PrP(C) physiology. To gain insights into the mechanisms of alpha-cleavage, we generated a library of PrP(C) mutants with mutations in the region neighbouring the alpha-cleavage site. The prevalence of C1, the carboxy adduct of alpha-cleavage, was determined for each mutant. In cell lines of disparate origin, C1 prevalence was unaffected by variations in charge and hydrophobicity of the region neighbouring the alpha-cleavage site, and by substitutions of the residues in the palindrome that flanks this site. Instead, alpha-cleavage was size-dependently impaired by deletions within the domain 106-119. Almost no cleavage was observed upon full deletion of this domain. These results suggest that alpha-cleavage is executed by an alpha-PrPase whose activity, despite surprisingly limited sequence specificity, is dependent on the size of the central region of PrP(C).
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.