1
|
Mao X, Lu X, Liu Y, Wu H, Li B, Bi X. Exploring the mediating role of cerebrospinal fluid metabolites in the pathway from circulating inflammatory proteins to multiple sclerosis: A Mendelian randomization study. Mult Scler Relat Disord 2025; 98:106440. [PMID: 40245661 DOI: 10.1016/j.msard.2025.106440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disease in which inflammation plays a pivotal role in its pathogenesis. The inflammatory response is regulated by a complex network of cells and mediators, including circulating proteins such as cytokines and inflammatory mediators. Metabolomics is a powerful analytical approach that may provide diagnostic and therapeutic targets for MS. However, the causal effects of circulating inflammatory proteins and cerebrospinal fluid metabolites (CSFMs) on MS, as well as whether CSFMs act as mediators, remain unclear. OBJECTIVE In this study, we obtained data on circulating inflammatory proteins, CSFMs, and MS from the largest genome-wide association study (GWAS) dataset of the International Multiple Sclerosis Genetics Consortium (IMSGC). METHODS We utilized the Mendelian randomization (MR) mediation analysis method to investigate the causal relationships among circulating inflammatory proteins, CSFMs and MS. Inverse variance weighting (IVW) served as the primary statistical method. Additionally, we explored whether CSFMs act as mediators in the pathway from circulating inflammatory proteins to MS. RESULTS Our findings reveal that there are five inflammatory proteins associated with MS. MR analysis reveals a positive correlation between the genetic prediction of three inflammatory proteins and the occurrence of MS. Our study reveals a link between 10 CSFMs and MS. Further MR analysis reveals a positive correlation between the genetic prediction of 6 CSFMs and the development of MS. Notably, CSFMs do not exhibit a reverse effect on MS. Our study establishes a significant causal effect of circulating inflammatory proteins and CSFMs on the progression of MS. Furthermore, CSFMs do not serve as an intermediary factor in the pathway connecting inflammatory proteins with MS. Circulating inflammatory proteins and CSFMs are causally associated with MS, and CSFMs do not appear to be intermediate factors in the pathway from inflammatory proteins to MS.
Collapse
Affiliation(s)
- Xiaowei Mao
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China
| | - Xiaoyan Lu
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China
| | - Yanqun Liu
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China
| | - Hangfei Wu
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China
| | - Binghan Li
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China.
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, Naval Medical University, 168 Changhai Road, Yangpu District, 200433, Shanghai, China.
| |
Collapse
|
2
|
Stojkovic L, Djordjevic A, Stefanovic M, Stankovic A, Dincic E, Djuric T, Zivkovic M. Circulatory Indicators of Lipid Peroxidation, the Driver of Ferroptosis, Reflect Differences between Relapsing-Remitting and Progressive Multiple Sclerosis. Int J Mol Sci 2024; 25:11024. [PMID: 39456806 PMCID: PMC11507982 DOI: 10.3390/ijms252011024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis, a lipid peroxidation- and iron-mediated type of regulated cell death, relates to both neuroinflammation, which is common in relapsing-remitting multiple sclerosis (RRMS), and neurodegeneration, which is prevalent in progressive (P)MS. Currently, findings related to the molecular markers proposed in this paper in patients are scarce. We analyzed circulatory molecular indicators of the main ferroptosis-related processes, comprising lipid peroxidation (malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and hexanoyl-lysine adduct (HEL)), glutathione-related antioxidant defense (total glutathione (reduced (GSH) and oxidized (GSSG)) and glutathione peroxidase 4 (GPX4)), and iron metabolism (iron, transferrin and ferritin) to estimate their contributions to the clinical manifestation of MS and differences between RRMS and PMS disease course. In 153 patients with RRMS and 69 with PMS, plasma/serum lipid peroxidation indicators and glutathione were quantified using ELISA and colorimetric reactions, respectively. Iron serum concentrations were determined using spectrophotometry, and transferrin and ferritin were determined using immunoturbidimetry. Compared to those with RRMS, patients with PMS had decreased 4-HNE (median, 1368.42 vs. 1580.17 pg/mL; p = 0.03). Interactive effects of MS course (RRMS/PMS) and disease-modifying therapy status on MDA (p = 0.009) and HEL (p = 0.02) levels were detected. In addition, the interaction of disease course and self-reported fatigue revealed significant impacts on 4-HNE levels (p = 0.01) and the GSH/GSSG ratio (p = 0.04). The results also show an association of MS course (p = 0.03) and EDSS (p = 0.04) with GSH levels. No significant changes were observed in the serum concentrations of iron metabolism indicators between the two patient groups (p > 0.05). We suggest circulatory 4-HNE as an important parameter related to differences between RRMS and PMS. Significant interactions of MS course and other clinically relevant parameters with changes in redox processes associated with ferroptosis support the further investigation of MS with a larger sample while taking into account both circulatory and central nervous system estimation.
Collapse
Affiliation(s)
- Ljiljana Stojkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Milan Stefanovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Evica Dincic
- Clinic for Neurology, Military Medical Academy, 11000 Belgrade, Serbia;
- Medical Faculty of the Military Medical Academy, University of Defence, 11000 Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| |
Collapse
|
3
|
Alwahsh M, Nimer RM, Dahabiyeh LA, Hamadneh L, Hasan A, Alejel R, Hergenröder R. NMR-based metabolomics identification of potential serum biomarkers of disease progression in patients with multiple sclerosis. Sci Rep 2024; 14:14806. [PMID: 38926483 PMCID: PMC11208524 DOI: 10.1038/s41598-024-64490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic and progressive neurological disorder, characterized by neuroinflammation and demyelination within the central nervous system (CNS). The etiology and the pathogenesis of MS are still unknown. Till now, no satisfactory treatments, diagnostic and prognostic biomarkers are available for MS. Therefore, we aimed to investigate metabolic alterations in patients with MS compared to controls and across MS subtypes. Metabolic profiles of serum samples from patients with MS (n = 90) and healthy control (n = 30) were determined by Nuclear Magnetic Resonance (1H-NMR) Spectroscopy using cryogenic probe. This approach was also utilized to identify significant differences between the metabolite profiles of the MS groups (primary progressive, secondary progressive, and relapsing-remitting) and the healthy controls. Concentrations of nine serum metabolites (adenosine triphosphate (ATP), tryptophan, formate, succinate, glutathione, inosine, histidine, pantothenate, and nicotinamide adenine dinucleotide (NAD)) were significantly higher in patients with MS compared to control. SPMS serum exhibited increased pantothenate and tryptophan than in PPMS. In addition, lysine, myo-inositol, and glutamate exhibited the highest discriminatory power (0.93, 95% CI 0.869-0.981; 0.92, 95% CI 0.859-0.969; 0.91, 95% CI 0.843-0.968 respectively) between healthy control and MS. Using NMR- based metabolomics, we identified a set of metabolites capable of classifying MS patients and controls. These findings confirmed untargeted metabolomics as a useful approach for the discovery of possible novel biomarkers that could aid in the diagnosis of the disease.
Collapse
Affiliation(s)
- Mohammad Alwahsh
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan.
| | - Refat M Nimer
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Lina A Dahabiyeh
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Lama Hamadneh
- Department of Badic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Aya Hasan
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan
| | - Rahaf Alejel
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 17138, Jordan
| | - Roland Hergenröder
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V., 44139, Dortmund, Germany
| |
Collapse
|
4
|
Xu D, Dai X, Zhang L, Cai Y, Chen K, Wu J, Dong L, Shen L, Yang J, Zhao J, Zhou Y, Mei Z, Wei W, Zhang Z, Xiong N. Mass spectrometry for biomarkers, disease mechanisms, and drug development in cerebrospinal fluid metabolomics. Trends Analyt Chem 2024; 173:117626. [DOI: 10.1016/j.trac.2024.117626] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
|
5
|
Židó M, Kačer D, Valeš K, Zimová D, Štětkářová I. Metabolomics of Cerebrospinal Fluid Amino and Fatty Acids in Early Stages of Multiple Sclerosis. Int J Mol Sci 2023; 24:16271. [PMID: 38003464 PMCID: PMC10671192 DOI: 10.3390/ijms242216271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating and neurodegenerative autoimmune disease of the central nervous system (CNS) damaging myelin and axons. Diagnosis is based on the combination of clinical findings, magnetic resonance imaging (MRI) and analysis of cerebrospinal fluid (CSF). Metabolomics is a systematic study that allows us to track amounts of different metabolites in a chosen medium. The aim of this study was to establish metabolomic differences between the cerebrospinal fluid of patients in the early stages of multiple sclerosis and healthy controls, which could potentially serve as markers for predicting disease activity. We collected CSF from 40 patients after the first attack of clinical symptoms who fulfilled revised McDonald criteria of MS, and the CSF of 33 controls. Analyses of CSF samples were performed by using the high-performance liquid chromatography system coupled with a mass spectrometer with a high-resolution detector. Significant changes in concentrations of arginine, histidine, spermidine, glutamate, choline, tyrosine, serine, oleic acid, stearic acid and linoleic acid were observed. More prominently, Expanded Disability Status Scale values significantly correlated with lower concentrations of histidine. We conclude that these metabolites could potentially play a role as a biomarker of disease activity and predict presumable inflammatory changes.
Collapse
Affiliation(s)
- Michal Židó
- Department of Neurology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
- Department of Neurology, Faculty Hospital Královské Vinohrady, 100 34 Prague, Czech Republic;
| | - David Kačer
- National Institute of Mental Health, 250 67 Klecany, Czech Republic; (D.K.); (K.V.)
| | - Karel Valeš
- National Institute of Mental Health, 250 67 Klecany, Czech Republic; (D.K.); (K.V.)
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic
| | - Denisa Zimová
- Department of Neurology, Faculty Hospital Královské Vinohrady, 100 34 Prague, Czech Republic;
| | - Ivana Štětkářová
- Department of Neurology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
- Department of Neurology, Faculty Hospital Královské Vinohrady, 100 34 Prague, Czech Republic;
| |
Collapse
|
6
|
Herman S, Arvidsson McShane S, Zjukovskaja C, Khoonsari PE, Svenningsson A, Burman J, Spjuth O, Kultima K. Disease phenotype prediction in multiple sclerosis. iScience 2023; 26:106906. [PMID: 37332601 PMCID: PMC10275960 DOI: 10.1016/j.isci.2023.106906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/09/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Progressive multiple sclerosis (PMS) is currently diagnosed retrospectively. Here, we work toward a set of biomarkers that could assist in early diagnosis of PMS. A selection of cerebrospinal fluid metabolites (n = 15) was shown to differentiate between PMS and its preceding phenotype in an independent cohort (AUC = 0.93). Complementing the classifier with conformal prediction showed that highly confident predictions could be made, and that three out of eight patients developing PMS within three years of sample collection were predicted as PMS at that time point. Finally, this methodology was applied to PMS patients as part of a clinical trial for intrathecal treatment with rituximab. The methodology showed that 68% of the patients decreased their similarity to the PMS phenotype one year after treatment. In conclusion, the inclusion of confidence predictors contributes with more information compared to traditional machine learning, and this information is relevant for disease monitoring.
Collapse
Affiliation(s)
- Stephanie Herman
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Payam Emami Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Box 1031, 17121 Solna, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Burman
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Shi T, Browne RW, Tamaño-Blanco M, Jakimovski D, Weinstock-Guttman B, Zivadinov R, Ramanathan M, Blair RH. Metabolomic profiles in relapsing-remitting and progressive multiple sclerosis compared to healthy controls: a five-year follow-up study. Metabolomics 2023; 19:44. [PMID: 37079261 DOI: 10.1007/s11306-023-02010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION AND OBJECTIVES Multiple sclerosis (MS) is a disease of the central nervous system associated with immune dysfunction, demyelination, and neurodegeneration. The disease has heterogeneous clinical phenotypes such as relapsing-remitting MS (RRMS) and progressive multiple sclerosis (PMS), each with unique pathogenesis. Metabolomics research has shown promise in understanding the etiologies of MS disease. However, there is a paucity of clinical studies with follow-up metabolomics analyses. This 5-year follow-up (5YFU) cohort study aimed to investigate the metabolomics alterations over time between different courses of MS patients and healthy controls and provide insights into metabolic and physiological mechanisms of MS disease progression. METHODS A cohort containing 108 MS patients (37 PMS and 71 RRMS) and 42 controls were followed up for a median of 5 years. Liquid chromatography-mass spectrometry (LC-MS) was applied for untargeted metabolomics profiling of serum samples of the cohort at both baseline and 5YFU. Univariate analyses with mixed-effect ANCOVA models, clustering, and pathway enrichment analyses were performed to identify patterns of metabolites and pathway changes across the time effects and patient groups. RESULTS AND CONCLUSIONS Out of 592 identified metabolites, the PMS group exhibited the most changes, with 219 (37%) metabolites changed over time and 132 (22%) changed within the RRMS group (Bonferroni adjusted P < 0.05). Compared to the baseline, there were more significant metabolite differences detected between PMS and RRMS classes at 5YFU. Pathway enrichment analysis detected seven pathways perturbed significantly during 5YFU in MS groups compared to controls. PMS showed more pathway changes compared to the RRMS group.
Collapse
Affiliation(s)
- Tiange Shi
- Department of Biostatistics, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Richard W Browne
- Department of Biotechnical and Laboratory Sciences, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Miriam Tamaño-Blanco
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Dejan Jakimovski
- Department of Neurology, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Department of Neurology, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Robert Zivadinov
- Department of Neurology, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
- Department of Neurology, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Rachael H Blair
- Department of Biostatistics, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA.
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
8
|
Ponce de Leon-Sanchez ER, Dominguez-Ramirez OA, Herrera-Navarro AM, Rodriguez-Resendiz J, Paredes-Orta C, Mendiola-Santibañez JD. A Deep Learning Approach for Predicting Multiple Sclerosis. MICROMACHINES 2023; 14:749. [PMID: 37420982 DOI: 10.3390/mi14040749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 07/09/2023]
Abstract
This paper proposes a deep learning model based on an artificial neural network with a single hidden layer for predicting the diagnosis of multiple sclerosis. The hidden layer includes a regularization term that prevents overfitting and reduces the model complexity. The purposed learning model achieved higher prediction accuracy and lower loss than four conventional machine learning techniques. A dimensionality reduction method was used to select the most relevant features from 74 gene expression profiles for training the learning models. The analysis of variance test was performed to identify the statistical difference between the mean of the proposed model and the compared classifiers. The experimental results show the effectiveness of the proposed artificial neural network.
Collapse
Affiliation(s)
| | - Omar Arturo Dominguez-Ramirez
- Centro de Investigación en Tecnologías de Información y Sistemas, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Mexico
| | | | | | | | | |
Collapse
|
9
|
Metabolomic Changes in Patients Affected by Multiple Sclerosis and Treated with Fingolimod. Metabolites 2023; 13:metabo13030428. [PMID: 36984868 PMCID: PMC10056460 DOI: 10.3390/metabo13030428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
Current treatment for Multiple Sclerosis (MS) consists of a multidisciplinary approach including disease-modifying therapies. The response to treatment is heterogeneous, representing a crucial challenge in the classification of patients. Metabolomics is an innovative tool that can identifies biomarkers/predictors of treatment response. We aimed to evaluate plasma metabolic changes in a group of naïve Relapsing-Remitting MS patients starting Fingolimod treatment, to find specific metabolomic features that predict the therapeutic response as well as the possible side effects. The study included 42 Relapsing-Remitting MS blood samples, of which 30 were classified as responders after two years of FINGO treatment, whereas 12 patients were Not-Responders. For fifteen patients, samples were collected at four time points: before starting the therapy; at six months after the initiation; at twelve months after; and at twenty-four months after initiation. The serum was analysed through Nuclear Magnetic Resonance and multivariate and univariate statistical analysis. Considering the single comparison between each time point, it was possible to identify a set of metabolites changing their concentrations based on the drug intake. FINGO influences aminoacidic and energy metabolisms and reduces oxidative stress and the activity of the immune system, both typical features of MS.
Collapse
|
10
|
Lorefice L, Pitzalis M, Murgia F, Fenu G, Atzori L, Cocco E. Omics approaches to understanding the efficacy and safety of disease-modifying treatments in multiple sclerosis. Front Genet 2023; 14:1076421. [PMID: 36793897 PMCID: PMC9922720 DOI: 10.3389/fgene.2023.1076421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
From the perspective of precision medicine, the challenge for the future is to improve the accuracy of diagnosis, prognosis, and prediction of therapeutic responses through the identification of biomarkers. In this framework, the omics sciences (genomics, transcriptomics, proteomics, and metabolomics) and their combined use represent innovative approaches for the exploration of the complexity and heterogeneity of multiple sclerosis (MS). This review examines the evidence currently available on the application of omics sciences to MS, analyses the methods, their limitations, the samples used, and their characteristics, with a particular focus on biomarkers associated with the disease state, exposure to disease-modifying treatments (DMTs), and drug efficacies and safety profiles.
Collapse
Affiliation(s)
- Lorena Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- *Correspondence: Lorena Lorefice,
| | - Maristella Pitzalis
- Institute for Genetic and Biomedical Research, National Research Council, Cagliari, Italy
| | - Federica Murgia
- Dpt of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giuseppe Fenu
- Department of Neurosciences, ARNAS Brotzu, Cagliari, Italy
| | - Luigi Atzori
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Center, Binaghi Hospital, ASL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
11
|
Genchi A, Brambilla E, Sangalli F, Radaelli M, Bacigaluppi M, Furlan R, Andolfo A, Drago D, Magagnotti C, Scotti GM, Greco R, Vezzulli P, Ottoboni L, Bonopane M, Capilupo D, Ruffini F, Belotti D, Cabiati B, Cesana S, Matera G, Leocani L, Martinelli V, Moiola L, Vago L, Panina-Bordignon P, Falini A, Ciceri F, Uglietti A, Sormani MP, Comi G, Battaglia MA, Rocca MA, Storelli L, Pagani E, Gaipa G, Martino G. Neural stem cell transplantation in patients with progressive multiple sclerosis: an open-label, phase 1 study. Nat Med 2023; 29:75-85. [PMID: 36624312 PMCID: PMC9873560 DOI: 10.1038/s41591-022-02097-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/17/2022] [Indexed: 01/11/2023]
Abstract
Innovative pro-regenerative treatment strategies for progressive multiple sclerosis (PMS), combining neuroprotection and immunomodulation, represent an unmet need. Neural precursor cells (NPCs) transplanted in animal models of multiple sclerosis have shown preclinical efficacy by promoting neuroprotection and remyelination by releasing molecules sustaining trophic support and neural plasticity. Here we present the results of STEMS, a prospective, therapeutic exploratory, non-randomized, open-label, single-dose-finding phase 1 clinical trial ( NCT03269071 , EudraCT 2016-002020-86), performed at San Raffaele Hospital in Milan, Italy, evaluating the feasibility, safety and tolerability of intrathecally transplanted human fetal NPCs (hfNPCs) in 12 patients with PMS (with evidence of disease progression, Expanded Disability Status Scale ≥6.5, age 18-55 years, disease duration 2-20 years, without any alternative approved therapy). The safety primary outcome was reached, with no severe adverse reactions related to hfNPCs at 2-year follow-up, clearly demonstrating that hfNPC therapy in PMS is feasible, safe and tolerable. Exploratory secondary analyses showed a lower rate of brain atrophy in patients receiving the highest dosage of hfNPCs and increased cerebrospinal fluid levels of anti-inflammatory and neuroprotective molecules. Although preliminary, these results support the rationale and value of future clinical studies with the highest dose of hfNPCs in a larger cohort of patients.
Collapse
Affiliation(s)
- Angela Genchi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Elena Brambilla
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sangalli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Radaelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bacigaluppi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Roberto Furlan
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annapaola Andolfo
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Denise Drago
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Magagnotti
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Maria Scotti
- grid.18887.3e0000000417581884Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Greco
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Vezzulli
- grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Linda Ottoboni
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bonopane
- grid.18887.3e0000000417581884Clinical Trial Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Capilupo
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ruffini
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Belotti
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Stefania Cesana
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Giada Matera
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Letizia Leocani
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Vittorio Martinelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Panina-Bordignon
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Falini
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Uglietti
- grid.414818.00000 0004 1757 8749Department of Gynaecology, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Pia Sormani
- grid.5606.50000 0001 2151 3065Biostatistics Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Giancarlo Comi
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | | | - Maria A. Rocca
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Gaipa
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
12
|
Disease Activity and Progression in Multiple Sclerosis: New Evidences and Future Perspectives. J Clin Med 2022; 11:jcm11226643. [PMID: 36431119 PMCID: PMC9695838 DOI: 10.3390/jcm11226643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, debilitating, autoimmune-mediated, inflammatory disease of the central nervous system (CNS), in which a combination of inflammation, demyelination and axonal degeneration takes place with extreme highly interpersonal variability [...].
Collapse
|
13
|
Gagliano A, Murgia F, Capodiferro AM, Tanca MG, Hendren A, Falqui SG, Aresti M, Comini M, Carucci S, Cocco E, Lorefice L, Roccella M, Vetri L, Sotgiu S, Zuddas A, Atzori L. 1H-NMR-Based Metabolomics in Autism Spectrum Disorder and Pediatric Acute-Onset Neuropsychiatric Syndrome. J Clin Med 2022; 11:6493. [PMID: 36362721 PMCID: PMC9658067 DOI: 10.3390/jcm11216493] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/03/2023] Open
Abstract
We recently described a unique plasma metabolite profile in subjects with pediatric acute-onset neuropsychiatric syndrome (PANS), suggesting pathogenic models involving specific patterns of neurotransmission, neuroinflammation, and oxidative stress. Here, we extend the analysis to a group of patients with autism spectrum disorder (ASD), as a consensus has recently emerged around its immune-mediated pathophysiology with a widespread involvement of brain networks. This observational case-control study enrolled patients referred for PANS and ASD from June 2019 to May 2020, as well as neurotypical age and gender-matched control subjects. Thirty-four PANS outpatients, fifteen ASD outpatients, and twenty-five neurotypical subjects underwent physical and neuropsychiatric evaluations, alongside serum metabolomic analysis with 1H-NMR. In supervised models, the metabolomic profile of ASD was significantly different from controls (p = 0.0001), with skewed concentrations of asparagine, aspartate, betaine, glycine, lactate, glucose, and pyruvate. Metabolomic separation was also observed between PANS and ASD subjects (p = 0.02), with differences in the concentrations of arginine, aspartate, betaine, choline, creatine phosphate, glycine, pyruvate, and tryptophan. We confirmed a unique serum metabolomic profile of PANS compared with both ASD and neurotypical subjects, distinguishing PANS as a pathophysiological entity per se. Tryptophan and glycine appear as neuroinflammatory fingerprints of PANS and ASD, respectively. In particular, a reduction in glycine would primarily affect NMDA-R excitatory tone, overall impairing downstream glutamatergic, dopaminergic, and GABAergic transmissions. Nonetheless, we found metabolomic similarities between PANS and ASD that suggest a putative role of N-methyl-D-aspartate receptor (NMDA-R) dysfunction in both disorders. Metabolomics-based approaches could contribute to the identification of novel ASD and PANS biomarkers.
Collapse
Affiliation(s)
- Antonella Gagliano
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
- Department of Health Science, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Murgia
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Agata Maria Capodiferro
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Marcello Giuseppe Tanca
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Aran Hendren
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Stella Giulia Falqui
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Michela Aresti
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Martina Comini
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Sara Carucci
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Regional Center, ASSL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09126 Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Regional Center, ASSL Cagliari, 09126 Cagliari, Italy
| | - Michele Roccella
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90128 Palermo, Italy
| | - Luigi Vetri
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Stefano Sotgiu
- Child Neuropsychiatry Unit, Department of Medicine, Surgery and Farmacy, University of Sassari, 07100 Sassari, Italy
| | - Alessandro Zuddas
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| |
Collapse
|
14
|
Systematic Review of NMR-Based Metabolomics Practices in Human Disease Research. Metabolites 2022; 12:metabo12100963. [PMID: 36295865 PMCID: PMC9609461 DOI: 10.3390/metabo12100963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 12/02/2022] Open
Abstract
Nuclear magnetic resonance (NMR) spectroscopy is one of the principal analytical techniques for metabolomics. It has the advantages of minimal sample preparation and high reproducibility, making it an ideal technique for generating large amounts of metabolomics data for biobanks and large-scale studies. Metabolomics is a popular “omics” technology and has established itself as a comprehensive exploratory biomarker tool; however, it has yet to reach its collaborative potential in data collation due to the lack of standardisation of the metabolomics workflow seen across small-scale studies. This systematic review compiles the different NMR metabolomics methods used for serum, plasma, and urine studies, from sample collection to data analysis, that were most popularly employed over a two-year period in 2019 and 2020. It also outlines how these methods influence the raw data and the downstream interpretations, and the importance of reporting for reproducibility and result validation. This review can act as a valuable summary of NMR metabolomic workflows that are actively used in human biofluid research and will help guide the workflow choice for future research.
Collapse
|
15
|
Liu Z, Waters J, Rui B. Metabolomics as a promising tool for improving understanding of multiple sclerosis: A review of recent advances. Biomed J 2022; 45:594-606. [PMID: 35042018 PMCID: PMC9486246 DOI: 10.1016/j.bj.2022.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 12/29/2021] [Accepted: 01/10/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system that usually affects young adults. The development of MS is closely related to the changes in the metabolome. Metabolomics studies have been performed using biofluids or tissue samples from rodent models and human patients to reveal metabolic alterations associated with MS progression. This review aims to provide an overview of the applications of metabolomics that for the investigations of the perturbed metabolic pathways in MS and to reveal the potential of metabolomics in personalizing treatments. In conclusion, informative variations of metabolites can be potential biomarkers in advancing our understanding of MS pathogenesis for MS diagnosis, predicting the progression of the disease, and estimating drug effects. Metabolomics will be a promising technique for improving clinical care in MS.
Collapse
Affiliation(s)
- Zhicheng Liu
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs School of Pharmacy, Anhui Medical University, Hefei, China.
| | - Jeffrey Waters
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Bin Rui
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA.
| |
Collapse
|
16
|
Weinstein DR, Owens GM, Gandhi A. Multiple Sclerosis: Systemic Challenges to Cost-Effective Care. AMERICAN HEALTH & DRUG BENEFITS 2022; 15:13-20. [PMID: 35586614 PMCID: PMC9038003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/22/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a progressive autoimmune disorder of the central nervous system characterized by symptoms including reduced mobility, pain, fatigue, and spasticity. MS affects nearly 1 million people in the United States, with significant negative impact on a patient's quality of life, and an average lifetime cost of care in excess of $4 million. The cost-effective management of patients with MS faces several challenges. OBJECTIVE To review the challenges to the cost-effective management of patients with MS, and to offer healthcare stakeholders a roadmap to address them. DISCUSSION The cost-effective management of patients with MS, which is driven largely by how quickly a patient receives effective medication therapy, is challenged by a paucity of between-office-visit clinical data, variability of provider expertise with magnetic resonance imaging (MRI), MRI machine quality, lack of standards for MRI machines and reports, misaligned financial incentives, the limited number of available Current Procedural Terminology (CPT) codes for brain MRI, the complexity of disease-modifying therapy (DMT) selection, poor patient adherence to treatment plans, poor communication among providers, and a lack of objective measures of disease progression. CONCLUSION Insurers, neurologists, researchers, and patient advocacy groups must address the needs of patients with MS holistically. These efforts should include establishing standards for MRI machines and reports, matching patients with MS specialists, aligning financial incentives, including creating a new CPT code for complex brain MRI, streamlining prior authorization processes of DMTs, using technology to gather patient data and improve coordination of care, and developing better measurement tools of disease activity.
Collapse
Affiliation(s)
| | - Gary M Owens
- President, Gary Owens Associates, Ocean View, DE
| | - Ankit Gandhi
- Clinical Trials Results Analyst, National Institutes of Health, Bethesda, MD
| |
Collapse
|
17
|
Toczylowska B, Zieminska E, Podlecka-Pietowska A, Ruszczynska A, Chalimoniuk M. Serum metabolic profiles and metal levels of patients with multiple sclerosis and patients with neuromyelitis optica spectrum disorders - NMR spectroscopy and ICP–MS studies. Mult Scler Relat Disord 2022; 60:103672. [DOI: 10.1016/j.msard.2022.103672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/24/2022] [Accepted: 02/05/2022] [Indexed: 11/29/2022]
|
18
|
Multi-Omics Approach to Elucidate Cerebrospinal Fluid Changes in Dogs with Intervertebral Disc Herniation. Int J Mol Sci 2021; 22:ijms222111678. [PMID: 34769107 PMCID: PMC8583948 DOI: 10.3390/ijms222111678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Herniation of the intervertebral disc (IVDH) is the most common cause of neurological and intervertebral disc degeneration-related diseases. Since the disc starts to degenerate before it can be observed by currently available diagnostic methods, there is an urgent need for novel diagnostic approaches. To identify molecular networks and pathways which may play important roles in intervertebral disc herniation, as well as to reveal the potential features which could be useful for monitoring disease progression and prognosis, multi-omics profiling, including high-resolution liquid chromatography-mass spectrometry (LC-MS)-based metabolomics and tandem mass tag (TMT)-based proteomics was performed. Cerebrospinal fluid of nine dogs with IVDH and six healthy controls were used for the analyses, and an additional five IVDH samples were used for proteomic data validation. Furthermore, multi-omics data were integrated to decipher a complex interaction between individual omics layers, leading to an improved prediction model. Together with metabolic pathways related to amino acids and lipid metabolism and coagulation cascades, our integromics prediction model identified the key features in IVDH, namely the proteins follistatin Like 1 (FSTL1), secretogranin V (SCG5), nucleobindin 1 (NUCB1), calcitonin re-ceptor-stimulating peptide 2 precursor (CRSP2) and the metabolites N-acetyl-D-glucosamine and adenine, involved in neuropathic pain, myelination, and neurotransmission and inflammatory response, respectively. Their clinical application is to be further investigated. The utilization of a novel integrative interdisciplinary approach may provide new opportunities to apply innovative diagnostic and monitoring methods as well as improve treatment strategies and personalized care for patients with degenerative spinal disorders.
Collapse
|
19
|
Rispoli MG, Valentinuzzi S, De Luca G, Del Boccio P, Federici L, Di Ioia M, Digiovanni A, Grasso EA, Pozzilli V, Villani A, Chiarelli AM, Onofrj M, Wise RG, Pieragostino D, Tomassini V. Contribution of Metabolomics to Multiple Sclerosis Diagnosis, Prognosis and Treatment. Int J Mol Sci 2021; 22:11112. [PMID: 34681773 PMCID: PMC8541167 DOI: 10.3390/ijms222011112] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolomics-based technologies map in vivo biochemical changes that may be used as early indicators of pathological abnormalities prior to the development of clinical symptoms in neurological conditions. Metabolomics may also reveal biochemical pathways implicated in tissue dysfunction and damage and thus assist in the development of novel targeted therapeutics for neuroinflammation and neurodegeneration. Metabolomics holds promise as a non-invasive, high-throughput and cost-effective tool for early diagnosis, follow-up and monitoring of treatment response in multiple sclerosis (MS), in combination with clinical and imaging measures. In this review, we offer evidence in support of the potential of metabolomics as a biomarker and drug discovery tool in MS. We also use pathway analysis of metabolites that are described as potential biomarkers in the literature of MS biofluids to identify the most promising molecules and upstream regulators, and show novel, still unexplored metabolic pathways, whose investigation may open novel avenues of research.
Collapse
Affiliation(s)
- Marianna Gabriella Rispoli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Silvia Valentinuzzi
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giovanna De Luca
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Piero Del Boccio
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Federici
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Di Ioia
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Anna Digiovanni
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Eleonora Agata Grasso
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Valeria Pozzilli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Alessandro Villani
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Antonio Maria Chiarelli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Marco Onofrj
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Richard G. Wise
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Damiana Pieragostino
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Paediatrics, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Valentina Tomassini
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| |
Collapse
|
20
|
Mass spectrometry based metabolomics of volume-restricted in-vivo brain samples: Actual status and the way forward. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Shi Y, Ding Y, Li G, Wang L, Osman RA, Sun J, Qian L, Zheng G, Zhang G. Discovery of Novel Biomarkers for Diagnosing and Predicting the Progression of Multiple Sclerosis Using TMT-Based Quantitative Proteomics. Front Immunol 2021; 12:700031. [PMID: 34489947 PMCID: PMC8417809 DOI: 10.3389/fimmu.2021.700031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/05/2021] [Indexed: 01/18/2023] Open
Abstract
Objective Here, we aimed to identify protein biomarkers that could rapidly and accurately diagnose multiple sclerosis (MS) using a highly sensitive proteomic immunoassay. Methods Tandem mass tag (TMT) quantitative proteomic analysis was performed to determine the differentially expressed proteins (DEPs) in cerebrospinal fluid (CSF) samples collected from 10 patients with MS and 10 non-inflammatory neurological controls (NINCs). The DEPs were analyzed using bioinformatics tools, and the candidate proteins were validated using the ELISA method in another cohort comprising 160 samples (paired CSF and plasma of 40 patients with MS, CSF of 40 NINCs, and plasma of 40 healthy individuals). Receiver operating characteristic (ROC) curves were used to determine the diagnostic potential of this method. Results Compared to NINCs, we identified 83 CSF-specific DEPs out of a total of 343 proteins in MS patients. Gene ontology (GO) enrichment analysis revealed that these DEPs are mainly involved in platelet degranulation, negative regulation of proteolysis, and post-translational protein modification. Pathway enrichment analysis revealed that the complement and coagulation cascades, Ras signaling pathway, and PI3K-Akt signaling pathway are the main components. Insulin-like growth factor-binding protein 7 (IGFBP7), insulin-like growth factor 2 (IGF2), and somatostatin (SST) were identified as the potential proteins with high scores, degree, and centrality in the protein-protein interaction (PPI) network. We validated the expression of these three proteins using ELISA. Compared to NINCs, the level of CSF IGFBP7 was significantly upregulated, and the level of CSF SST was significantly downregulated in the MS group. Conclusion Our results suggest that SST and IGFBP7 might be associated with the pathogenesis of MS and would be helpful in diagnosing MS. Since IGFBP7 was used to classify relapsing remitting MS (RRMS) and secondary progressive MS (SPMS) patients, therefore, it may act as a potential key marker and therapeutic target in MS.
Collapse
Affiliation(s)
- Yijun Shi
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaowei Ding
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guoge Li
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lijuan Wang
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,NMPA Key Laboratory for Quality Control of In Vitro Diagnostics , Beijing, China.,Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing, China
| | - Rasha Alsamani Osman
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jialu Sun
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lingye Qian
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guanghui Zheng
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,NMPA Key Laboratory for Quality Control of In Vitro Diagnostics , Beijing, China.,Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing, China
| | - Guojun Zhang
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,NMPA Key Laboratory for Quality Control of In Vitro Diagnostics , Beijing, China.,Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing, China
| |
Collapse
|
22
|
Pautova A, Burnakova N, Revelsky A. Metabolic Profiling and Quantitative Analysis of Cerebrospinal Fluid Using Gas Chromatography-Mass Spectrometry: Current Methods and Future Perspectives. Molecules 2021; 26:3597. [PMID: 34208377 PMCID: PMC8231178 DOI: 10.3390/molecules26123597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Cerebrospinal fluid is a key biological fluid for the investigation of new potential biomarkers of central nervous system diseases. Gas chromatography coupled to mass-selective detectors can be used for this investigation at the stages of metabolic profiling and method development. Different sample preparation conditions, including extraction and derivatization, can be applied for the analysis of the most of low-molecular-weight compounds of the cerebrospinal fluid, including metabolites of tryptophan, arachidonic acid, glucose; amino, polyunsaturated fatty and other organic acids; neuroactive steroids; drugs; and toxic metabolites. The literature data analysis revealed the absence of fully validated methods for cerebrospinal fluid analysis, and it presents opportunities for scientists to develop and validate analytical protocols using modern sample preparation techniques, such as microextraction by packed sorbent, dispersive liquid-liquid microextraction, and other potentially applicable techniques.
Collapse
Affiliation(s)
- Alisa Pautova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Laboratory of Human Metabolism in Critical States, Negovsky Research Institute of General Reanimatology, Petrovka str. 25-2, 107031 Moscow, Russia
| | - Natalia Burnakova
- Laboratory of Mass Spectrometry, Chemistry Department, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 1-3, 119991 Moscow, Russia; (N.B.); (A.R.)
| | - Alexander Revelsky
- Laboratory of Mass Spectrometry, Chemistry Department, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 1-3, 119991 Moscow, Russia; (N.B.); (A.R.)
| |
Collapse
|
23
|
Rivera-Velez SM, Navas J, Villarino NF. Applying metabolomics to veterinary pharmacology and therapeutics. J Vet Pharmacol Ther 2021; 44:855-869. [PMID: 33719079 DOI: 10.1111/jvp.12961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Metabolomics is the large-scale study of low-molecular-weight substances in a biological system in a given physiological state at a given time point. Metabolomics can be applied to identify predictors of inter-individual variability in drug response, provide clinicians with data useful for decision-making processes in drug selection, and inform about the pharmacokinetics and pharmacodynamics of a drug. It is, therefore, an exceptional approach for gaining new understanding effects in the field of comparative veterinary pharmacology. However, the incorporation of metabolomics into veterinary pharmacology and toxicology is not yet widespread, and this is probably, at least in part, a result of its highly multidisciplinary nature. This article reviews the potential applications of metabolomics in veterinary pharmacology and therapeutics. It integrates key concepts for designing metabolomics studies and analyzing and interpreting metabolomics data, providing solid foundations for applying metabolomics to the study of drugs in all veterinary species.
Collapse
Affiliation(s)
- Sol M Rivera-Velez
- Molecular Determinants Core, Johns Hopkins All Children's Hospital, Saint Petersburg, Florida, USA
| | - Jinna Navas
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nicolas F Villarino
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
24
|
Zahoor I, Rui B, Khan J, Datta I, Giri S. An emerging potential of metabolomics in multiple sclerosis: a comprehensive overview. Cell Mol Life Sci 2021; 78:3181-3203. [PMID: 33449145 PMCID: PMC8038957 DOI: 10.1007/s00018-020-03733-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/14/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the nervous system that primarily affects young adults. Although the exact etiology of the disease remains obscure, it is clear that alterations in the metabolome contribute to this process. As such, defining a reliable and disease-specific metabolome has tremendous potential as a diagnostic and therapeutic strategy for MS. Here, we provide an overview of studies aimed at identifying the role of metabolomics in MS. These offer new insights into disease pathophysiology and the contributions of metabolic pathways to this process, identify unique markers indicative of treatment responses, and demonstrate the therapeutic effects of drug-like metabolites in cellular and animal models of MS. By and large, the commonly perturbed pathways in MS and its preclinical model include lipid metabolism involving alpha-linoleic acid pathway, nucleotide metabolism, amino acid metabolism, tricarboxylic acid cycle, d-ornithine and d-arginine pathways with collective role in signaling and energy supply. The metabolomics studies suggest that metabolic profiling of MS patient samples may uncover biomarkers that will advance our understanding of disease pathogenesis and progression, reduce delays and mistakes in diagnosis, monitor the course of disease, and detect better drug targets, all of which will improve early therapeutic interventions and improve evaluation of response to these treatments.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA. .,Department of Neurology, Henry Ford Hospital, Education & Research Building, Room 4023, 2799 W Grand Blvd, Detroit, MI, 48202, USA.
| | - Bin Rui
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Junaid Khan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA. .,Department of Neurology, Henry Ford Hospital, Education & Research Building, Room 4051, 2799 W Grand Blvd, Detroit, MI, 48202, USA.
| |
Collapse
|
25
|
Murgia F, Atzori L, Carboni E, Santoru ML, Hendren A, Pisanu A, Caboni P, Boi L, Fusco G, Carta AR. Metabolomics Fingerprint Induced by the Intranigral Inoculation of Exogenous Human Alpha-Synuclein Oligomers in a Rat Model of Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21186745. [PMID: 32937957 PMCID: PMC7555976 DOI: 10.3390/ijms21186745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is considered a synucleinopathy because of the intraneuronal accumulation of aggregated α-synuclein (αSyn). Recent evidence points to soluble αSyn-oligomers (αSynO) as the main cytotoxic species responsible for cell death. Given the pivotal role of αSyn in PD, αSyn-based models are crucial for the investigation of toxic mechanisms and the identification of new therapeutic targets in PD. By using a metabolomics approach, we evaluated the metabolic profile of brain and serum samples of rats infused unilaterally with preformed human αSynOs (HαSynOs), or vehicle, into the substantia nigra pars compacta (SNpc). Three months postinfusion, the striatum was dissected for striatal dopamine (DA) measurements via High Pressure Liquid Chromatography (HPLC) analysis and mesencephalon and serum samples were collected for the evaluation of metabolite content via gas chromatography mass spectrometry analysis. Multivariate, univariate and correlation statistics were applied. A 40% decrease of DA content was measured in the HαSynO-infused striatum as compared to the contralateral and the vehicle-infused striata. Decreased levels of dehydroascorbic acid, myo-inositol, and glycine, and increased levels of threonine, were found in the mesencephalon, while increased contents of fructose and mannose, and a decrease in glycine and urea, were found in the serum of HαSynO-infused rats. The significant correlation between DA and metabolite content indicated that metabolic variations reflected the nigrostriatal degeneration. Collectively, the metabolomic fingerprint of HαSynO-infused rats points to an increase of oxidative stress markers, in line with PD neuropathology, and provides hints for potential biomarkers of PD.
Collapse
Affiliation(s)
- Federica Murgia
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (L.A.); (M.L.S.); (A.H.)
- Correspondence: (F.M.); (A.R.C.)
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (L.A.); (M.L.S.); (A.H.)
| | - Ezio Carboni
- Neuroscience Section, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (E.C.); (L.B.)
| | - Maria Laura Santoru
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (L.A.); (M.L.S.); (A.H.)
| | - Aran Hendren
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (L.A.); (M.L.S.); (A.H.)
- Faculty of Health and Medical Sciences, University of Surrey, London GU2 7XH, UK
| | - Augusta Pisanu
- CNR Institute of Neuroscience, Monserrato, 09042 Cagliari, Italy;
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy;
| | - Laura Boi
- Neuroscience Section, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (E.C.); (L.B.)
| | - Giuliana Fusco
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge CB2 1EW, UK;
| | - Anna R. Carta
- Neuroscience Section, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042 Cagliari, Italy; (E.C.); (L.B.)
- Correspondence: (F.M.); (A.R.C.)
| |
Collapse
|
26
|
Albrecht B, Voronina E, Schipke C, Peters O, Parr MK, Díaz-Hernández MD, Schlörer NE. Pursuing Experimental Reproducibility: An Efficient Protocol for the Preparation of Cerebrospinal Fluid Samples for NMR-based Metabolomics and Analysis of Sample Degradation. Metabolites 2020; 10:metabo10060251. [PMID: 32560109 PMCID: PMC7345835 DOI: 10.3390/metabo10060251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022] Open
Abstract
NMR-based metabolomics investigations of human biofluids offer great potential to uncover new biomarkers. In contrast to protocols for sample collection and biobanking, procedures for sample preparation prior to NMR measurements are still heterogeneous, thus compromising the comparability of the resulting data. Herein, we present results of an investigation of the handling of cerebrospinal fluid (CSF) samples for NMR metabolomics research. Origins of commonly observed problems when conducting NMR experiments on this type of sample are addressed, and suitable experimental conditions in terms of sample preparation and pH control are discussed. Sample stability was assessed by monitoring the degradation of CSF samples by NMR, hereby identifying metabolite candidates, which are potentially affected by sample storage. A protocol was devised yielding consistent spectroscopic data as well as achieving overall sample stability for robust analysis. We present easy to adopt standard operating procedures with the aim to establish a shared sample handling strategy that facilitates and promotes inter-laboratory comparison, and the analysis of sample degradation provides new insights into sample stability.
Collapse
Affiliation(s)
- Benjamin Albrecht
- Department of Chemistry, Universität zu Köln, Greinstr.4, 50939 Köln, Germany; (B.A.); (E.V.)
| | - Elena Voronina
- Department of Chemistry, Universität zu Köln, Greinstr.4, 50939 Köln, Germany; (B.A.); (E.V.)
| | - Carola Schipke
- Charité– Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Experimental & Clinical Research Center (ECRC), Lindenberger Weg 80, 13125 Berlin, Germany;
| | - Oliver Peters
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Maria Kristina Parr
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany;
| | - M. Dolores Díaz-Hernández
- Department of Chemistry, Universität zu Köln, Greinstr.4, 50939 Köln, Germany; (B.A.); (E.V.)
- Correspondence: (M.D.D.-H.); (N.E.S.); Tel.: +49-221-470-3081 (N.E.S.)
| | - Nils E. Schlörer
- Department of Chemistry, Universität zu Köln, Greinstr.4, 50939 Köln, Germany; (B.A.); (E.V.)
- Correspondence: (M.D.D.-H.); (N.E.S.); Tel.: +49-221-470-3081 (N.E.S.)
| |
Collapse
|