1
|
Sabaie H, Khorami Rouz S, Kouchakali G, Heydarzadeh S, Asadi MR, Sharifi-Bonab M, Hussen BM, Taheri M, Ayatollahi SA, Rezazadeh M. Identification of potential regulatory long non-coding RNA-associated competing endogenous RNA axes in periplaque regions in multiple sclerosis. Front Genet 2022; 13:1011350. [PMID: 36324503 PMCID: PMC9619104 DOI: 10.3389/fgene.2022.1011350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022] Open
Abstract
Slow-burning inflammation at the lesion rim is connected to the expansion of chronic multiple sclerosis (MS) lesions. However, the underlying processes causing expansion are not clearly realized. In this context, the current study used a bioinformatics approach to identify the expression profiles and related lncRNA-associated ceRNA regulatory axes in the periplaque region in MS patients. Expression data (GSE52139) from periplaque regions in the secondary progressive MS spinal cord and controls were downloaded from the Gene Expression Omnibus database (GEO), which has details on mRNAs and lncRNAs. Using the R software's limma package, the differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) were found. The RNA interactions were also found using the DIANA-LncBase, miRTarBase, and HMDD databases. The Pearson correlation coefficient was used to determine whether there were any positive correlations between DEmRNAs and DElncRNAs in the ceRNA network. Finally, lncRNA-associated ceRNA axes were created based on co-expression and connections between DElncRNA, miRNA, and DEmRNA. We used the Enrichr tool to enrich the biological process, molecular function, and pathways for DEmRNAs and DElncRNAs. A network of DEmRNAs' protein-protein interactions was developed, and the top five hub genes were found using Cytoscape and STRING. The current study indicates that 15 DEmRNAs, including FOS, GJA1, NTRK2, CTNND1, and SP3, are connected to the MS ceRNA network. Additionally, four DElncRNAs (such as TUG1, ASB16-AS1, and LINC01094) that regulated the aforementioned mRNAs by sponging 14 MS-related miRNAs (e.g., hsa-miR-145-5p, hsa-miR-200a-3p, hsa-miR-20a-5p, hsa-miR-22-3p, hsa-miR-23a-3p, hsa-miR-27a-3p, hsa-miR-29b-3p, hsa-miR-29c-3p, hsa-miR-34a-5p) were found. In addition, the analysis of pathway enrichment revealed that DEmRNAs were enriched in the pathways for the "MAPK signaling pathway", "Kaposi sarcoma-associated herpesvirus infection", "Human immunodeficiency virus one infection", "Lipid and atherosclerosis", and "Amphetamine addiction". Even though the function of these ceRNA axes needs to be investigated further, this study provides research targets for studying ceRNA-mediated molecular mechanisms related to periplaque demyelination in MS.
Collapse
Affiliation(s)
- Hani Sabaie
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ghazal Kouchakali
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Heydarzadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Asadi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mirmohsen Sharifi-Bonab
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | | | - Maryam Rezazadeh
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Fissolo N, Matute-Blanch C, Osman M, Costa C, Pinteac R, Miró B, Sanchez A, Brito V, Dujmovic I, Voortman M, Khalil M, Borràs E, Sabidó E, Issazadeh-Navikas S, Montalban X, Comabella Lopez M. CSF SERPINA3 Levels Are Elevated in Patients With Progressive MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/2/e941. [PMID: 33436375 PMCID: PMC8105904 DOI: 10.1212/nxi.0000000000000941] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Objective To identify biomarkers associated with progressive phases of MS and with neuroprotective potential. Methods Combined analysis of the transcriptional and proteomic profiles obtained in CNS tissue during chronic progressive phases of experimental autoimmune encephalomyelitis (EAE) with the transcriptional profile obtained during the differentiation of murine neural stem cells into neurons. Candidate biomarkers were measured by ELISA in the CSF of 65 patients with MS (29 with relapsing-remitting MS [RRMS], 20 with secondary progressive MS, and 16 with primary progressive MS [PPMS]) and 30 noninflammatory neurologic controls (NINCs). Results Integrative analysis of gene and protein expression data identified 2 biomarkers, the serine protease inhibitor Serpina3n and the calcium-binding protein S100A4, which were upregulated in chronic progressive EAE and whose expression was induced during neuronal differentiation. Immunofluorescence studies revealed a primarily neuronal expression of S100A4 and Serpina3n during EAE. CSF levels of SERPINA3, the human ortholog of murine Serpina3n, and S100A4 were increased in patients with MS compared with NINCs (SERPINA3: 1,320 vs 838.6 ng/mL, p = 0.0001; S100A4: 1.6 vs 0.8 ng/mL, p = 0.02). Within the MS group, CSF SERPINA3 levels were significantly elevated in patients with progressive forms, mainly patients with PPMS compared with patients with RRMS (1,617 vs 1,129 ng/mL, p = 0.02) and NINCs (1,617 vs 838.6 ng/mL, p = 0.0001). Of interest, CSF SERPINA3 levels significantly correlated with CSF neurofilament light chain levels only in the PPMS group (r = 0.62, p = 0.01). Conclusion These results point to a role of SERPINA3 as a biomarker associated with the progressive forms of MS, particularly PPMS.
Collapse
Affiliation(s)
- Nicolás Fissolo
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Clara Matute-Blanch
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Mohamoud Osman
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Carme Costa
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Rucsanda Pinteac
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Berta Miró
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Alex Sanchez
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Verónica Brito
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Irena Dujmovic
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Margarete Voortman
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Michael Khalil
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Eva Borràs
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Eduard Sabidó
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Shohreh Issazadeh-Navikas
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Xavier Montalban
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada
| | - Manuel Comabella Lopez
- From the Servei de Neurologia-Neuroimmunologia (N.F., C.M.-B., C.C., R.P., V.B., X.M., M.C.L.), Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Spain; Biotech Research and Innovation Centre (BRIC) (M.O., S.I.-N.), University of Copenhagen, Denmark; Statistics and Bioinformatics Unit (B.M., A.S.), Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain; Department of Neurology (I.D.), University of Belgrade School of Medicine, Serbia; Department of Neurology (I.D.), University of North Carolina School of Medicine, Chapel Hill; Department of Neurology (M.V., M.K.), Medical University of Graz, Austria; Proteomics Unit (E.B., E.S.), Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Spain; Proteomics Unit (E.B., E.S.), Universitat Pompeu Fabra, Barcelona, Spain; and Center for Multiple Sclerosis (X.M.), St. Michael's Hospital, University of Toronto, ON, Canada.
| |
Collapse
|
3
|
Autoantibodies against NMDA receptor 1 modify rather than cause encephalitis. Mol Psychiatry 2021; 26:7746-7759. [PMID: 34331009 PMCID: PMC8872987 DOI: 10.1038/s41380-021-01238-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023]
Abstract
The etiology and pathogenesis of "anti-N-methyl-D-aspartate-receptor (NMDAR) encephalitis" and the role of autoantibodies (AB) in this condition are still obscure. While NMDAR1-AB exert NMDAR-antagonistic properties by receptor internalization, no firm evidence exists to date that NMDAR1-AB by themselves induce brain inflammation/encephalitis. NMDAR1-AB of all immunoglobulin classes are highly frequent across mammals with multiple possible inducers and boosters. We hypothesized that "NMDAR encephalitis" results from any primary brain inflammation coinciding with the presence of NMDAR1-AB, which may shape the encephalitis phenotype. Thus, we tested whether following immunization with a "cocktail" of 4 NMDAR1 peptides, induction of a spatially and temporally defined sterile encephalitis by diphtheria toxin-mediated ablation of pyramidal neurons ("DTA" mice) would modify/aggravate the ensuing phenotype. In addition, we tried to replicate a recent report claiming that immunizing just against the NMDAR1-N368/G369 region induced brain inflammation. Mice after DTA induction revealed a syndrome comprising hyperactivity, hippocampal learning/memory deficits, prefrontal cortical network dysfunction, lasting blood brain-barrier impairment, brain inflammation, mainly in hippocampal and cortical regions with pyramidal neuronal death, microgliosis, astrogliosis, modest immune cell infiltration, regional atrophy, and relative increases in parvalbumin-positive interneurons. The presence of NMDAR1-AB enhanced the hyperactivity (psychosis-like) phenotype, whereas all other readouts were identical to control-immunized DTA mice. Non-DTA mice with or without NMDAR1-AB were free of any encephalitic signs. Replication of the reported NMDAR1-N368/G369-immunizing protocol in two large independent cohorts of wild-type mice completely failed. To conclude, while NMDAR1-AB can contribute to the behavioral phenotype of an underlying encephalitis, induction of an encephalitis by NMDAR1-AB themselves remains to be proven.
Collapse
|
4
|
Jäkel S, Williams A. What Have Advances in Transcriptomic Technologies Taught us About Human White Matter Pathologies? Front Cell Neurosci 2020; 14:238. [PMID: 32848627 PMCID: PMC7418269 DOI: 10.3389/fncel.2020.00238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/07/2020] [Indexed: 11/16/2022] Open
Abstract
For a long time, post-mortem analysis of human brain pathologies has been purely descriptive, limiting insight into the pathological mechanisms. However, starting in the early 2000s, next-generation sequencing (NGS) and the routine application of bulk RNA-sequencing and microarray technologies have revolutionized the usefulness of post-mortem human brain tissue. This has allowed many studies to provide novel mechanistic insights into certain brain pathologies, albeit at a still unsatisfying resolution, with masking of lowly expressed genes and regulatory elements in different cell types. The recent rapid evolution of single-cell technologies has now allowed researchers to shed light on human pathologies at a previously unreached resolution revealing further insights into pathological mechanisms that will open the way for the development of new strategies for therapies. In this review article, we will give an overview of the incremental information that single-cell technologies have given us for human white matter (WM) pathologies, summarize which single-cell technologies are available, and speculate where these novel approaches may lead us for pathological assessment in the future.
Collapse
Affiliation(s)
- Sarah Jäkel
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
5
|
Morris AH, Hughes KR, Oakes RS, Cai MM, Miller SD, Irani DN, Shea LD. Engineered immunological niches to monitor disease activity and treatment efficacy in relapsing multiple sclerosis. Nat Commun 2020; 11:3871. [PMID: 32747712 PMCID: PMC7398910 DOI: 10.1038/s41467-020-17629-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 07/09/2020] [Indexed: 12/19/2022] Open
Abstract
Relapses in multiple sclerosis can result in irreversible nervous system tissue injury. If these events could be detected early, targeted immunotherapy could potentially slow disease progression. We describe the use of engineered biomaterial-based immunological niches amenable to biopsy to provide insights into the phenotype of innate immune cells that control disease activity in a mouse model of multiple sclerosis. Differential gene expression in cells from these niches allow monitoring of disease dynamics and gauging the effectiveness of treatment. A proactive treatment regimen, given in response to signal within the niche but before symptoms appeared, substantially reduced disease. This technology offers a new approach to monitor organ-specific autoimmunity, and represents a platform to analyze immune dysfunction within otherwise inaccessible target tissues.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert S Oakes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michelle M Cai
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative autoimmune disease with a complex clinical course characterized by inflammation, demyelination, and axonal degeneration. Diagnosis of MS most commonly includes finding lesions in at least two separate areas of the central nervous system (CNS), including the brain, spinal cord, and optic nerves. In recent years, there has been a remarkable increase in the number of available treatments for MS. An optimal treatment is usually based on a personalized approach determined by an individual patient's prognosis and treatment risks. Biomarkers that can predict disability progression, monitor ongoing disease activity, and assess treatment response are integral in making important decisions regarding MS treatment. This review describes MS biomarkers that are currently being used in clinical practice; it also reviews and consolidates published findings from clinically relevant potential MS biomarkers in recent years. The work also discusses the challenges of validating and application of biomarkers in MS clinical practice.
Collapse
Affiliation(s)
- Anu Paul
- Department of Neurology, Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Manuel Comabella
- Department of Neurology, MS Centre of Catalonia, Vall d'Hebron University Hospital, Barcelona 08035, Spain
| | - Roopali Gandhi
- Department of Neurology, Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
7
|
Magro-Checa C, Steup-Beekman GM, Huizinga TW, van Buchem MA, Ronen I. Laboratory and Neuroimaging Biomarkers in Neuropsychiatric Systemic Lupus Erythematosus: Where Do We Stand, Where To Go? Front Med (Lausanne) 2018; 5:340. [PMID: 30564579 PMCID: PMC6288259 DOI: 10.3389/fmed.2018.00340] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/19/2018] [Indexed: 01/18/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by multi-systemic involvement. Nervous system involvement in SLE leads to a series of uncommon and heterogeneous neuropsychiatric (NP) manifestations. Current knowledge on the underlying pathogenic processes and their subsequent pathophysiological changes leading to NP-SLE manifestations is incomplete. Several putative laboratory biomarkers have been proposed as contributors to the genesis of SLE-related nervous system damage. Alongside the laboratory biomarkers, several neuroimaging tools have shown to reflect the nature of tissue microstructural damage associated with SLE, and thus were suggested to contribute to the understanding of the pathophysiological changes and subsequently help in clinical decision making. However, the number of useful biomarkers in NP-SLE in clinical practice is disconcertingly modest. In some cases it is not clear whether the biomarker is truly involved in pathogenesis, or the result of non-specific pathophysiological changes in the nervous system (e.g., neuroinflammation) or whether it is the consequence of a concomitant underlying abnormality related to SLE activity. In order to improve the diagnosis of NP-SLE and provide a better targeted care to these patients, there is still a need to develop and validate a range of biomarkers that reliably capture the different aspects of disease heterogeneity. This article critically reviews the current state of knowledge on laboratory and neuroimaging biomarkers in NP-SLE, discusses the factors that need to be addressed to make these biomarkers suitable for clinical application, and suggests potential future research paths to address important unmet needs in the NP-SLE field.
Collapse
Affiliation(s)
- César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands.,Department of Rheumatology, Zuyderland Medical Center, Heerlen, Netherlands
| | | | - Tom W Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands.,Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Itamar Ronen
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
8
|
Rajasundaram S. Adenosine A2A Receptor Signaling in the Immunopathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2018; 9:402. [PMID: 29559972 PMCID: PMC5845642 DOI: 10.3389/fimmu.2018.00402] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/13/2018] [Indexed: 12/19/2022] Open
Abstract
Our increasing appreciation of adenosine as an endogenous signaling molecule that terminates inflammation has generated excitement regarding the potential to target adenosine receptors (ARs) in the treatment of multiple sclerosis (MS), a disease of chronic neuroinflammation. Of the four G protein-coupled ARs, A2ARs are the principal mediator of adenosine’s anti-inflammatory effects and accordingly, there is a growing body of evidence surrounding the role of A2ARs in experimental autoimmune encephalomyelitis (EAE), the dominant animal model of MS. Such evidence points to a complex, often paradoxical role for A2ARs in the immunopathogenesis of EAE, where they have the ability to both exacerbate and alleviate disease severity. This review seeks to interpret these paradoxical findings and evaluate the therapeutic promise of A2ARs. In essence, the complexities of A2AR signaling arise from two properties. Firstly, A2AR signaling downregulates the inflammatory potential of TH lymphocytes whilst simultaneously facilitating the recruitment of these cells into the CNS. Secondly, A2AR expression by myeloid cells – infiltrating macrophages and CNS-resident microglia – has the capacity to promote both tissue injury and repair in chronic neuroinflammation. Consequently, the therapeutic potential of targeting A2ARs is greatly undermined by the risk of collateral tissue damage in the periphery and/or CNS.
Collapse
|
9
|
Shirvani-Farsani Z, Kakhki MP, Gargari BN, Doosti R, Moghadasi AN, Azimi AR, Behmanesh M. The expression of VDR mRNA but not NF-κB surprisingly decreased after vitamin D treatment in multiple sclerosis patients. Neurosci Lett 2017; 653:258-263. [PMID: 28576565 DOI: 10.1016/j.neulet.2017.05.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE The aim of this study was to investigate the expression levels of vitamin D receptor (VDR) and NF-κB mRNAs in vitamin D (VD) supplemented multiple sclerosis (MS) patients. METHODS RRMS patients received 50,000 IU vitamin D3/week as an intra-muscular injection for 2 months. Blood samples were obtained from 30 MS patients before and after VD supplementation and 32 healthy individuals, and then VDR and NF-κB mRNA levels were measured by real time PCR method and analyzed with independent and paired t-tests. Moreover, some correlations were performed between the expression levels of selected genes and some clinical features of MS and control groups. RESULTS Surprisingly, the expression level of VDR mRNA significantly decreased after 2 months supplementation with VD in our selected patients and in contrast, the level of serum 25(OH) D increased after supplementation. Although, we didn't find any significant difference in the expression level of NF-κB gene before and after treatment with VD, its expression significantly decreased in untreated MS cases compared with healthy controls. CONCLUSION In conclusion, we found some new evidences from the molecular mechanism of vitamin D effectiveness in MS treatment. Also, we need more functional studies to find the effect of VD on the expression level of VDR mRNA.
Collapse
Affiliation(s)
- Zeinab Shirvani-Farsani
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences and Technology, Shahid Beheshti University G.C., Tehran, Iran
| | - Majid Pahlevan Kakhki
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahar Naghavi Gargari
- Department of Basic Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roozita Doosti
- MS Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | | | - Amir Reza Azimi
- MS Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Cordiglieri C, Baggi F, Bernasconi P, Kapetis D, Faggiani E, Consonni A, Andreetta F, Frangiamore R, Confalonieri P, Antozzi C, Mantegazza R. Identification of a gene expression signature in peripheral blood of multiple sclerosis patients treated with disease-modifying therapies. Clin Immunol 2016; 173:133-146. [PMID: 27720845 DOI: 10.1016/j.clim.2016.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/30/2016] [Accepted: 10/04/2016] [Indexed: 01/17/2023]
Abstract
Multiple Sclerosis (MS) is an inflammatory disease with neurodegenerative alterations, ultimately progressing to neurological handicap. Therapies are effective in counteracting inflammation but not neurodegeneration. Biomarkers predicting disease course or treatment response are lacking. We investigated whether altered gene and protein expression profiles were detectable in the peripheral blood of 78 relapsing remitting MS (RR-MS) patients treated by disease-modifying therapies. A discovery/validation study on RR-MS responsive to glatiramer acetate identified 8 differentially expressed genes: ITGA2B, ITGB3, CD177, IGJ, IL5RA, MMP8, P2RY12, and S100β. A longitudinal study on glatiramer acetate, Interferon-β, or Fingolimod treated RR-MS patients confirmed that 7 out of 8 genes were downregulated with reference to the different therapies, whereas S100β was always upregulated. Thus, we identified a peripheral gene signature associated with positive response in RR-MS which may also explain drug immunomodulatory effects. The usefulness of this signature as a biomarker needs confirmation on larger series of patients.
Collapse
Affiliation(s)
- Chiara Cordiglieri
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Pia Bernasconi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Dimos Kapetis
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Elisa Faggiani
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Alessandra Consonni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Francesca Andreetta
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Rita Frangiamore
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Paolo Confalonieri
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Carlo Antozzi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute "Carlo Besta", Milan, Italy.
| |
Collapse
|
11
|
Wheeler NA, Fuss B. Extracellular cues influencing oligodendrocyte differentiation and (re)myelination. Exp Neurol 2016; 283:512-30. [PMID: 27016069 PMCID: PMC5010977 DOI: 10.1016/j.expneurol.2016.03.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
There is an increasing number of neurologic disorders found to be associated with loss and/or dysfunction of the CNS myelin sheath, ranging from the classic demyelinating disease, multiple sclerosis, through CNS injury, to neuropsychiatric diseases. The disabling burden of these diseases has sparked a growing interest in gaining a better understanding of the molecular mechanisms regulating the differentiation of the myelinating cells of the CNS, oligodendrocytes (OLGs), and the process of (re)myelination. In this context, the importance of the extracellular milieu is becoming increasingly recognized. Under pathological conditions, changes in inhibitory as well as permissive/promotional cues are thought to lead to an overall extracellular environment that is obstructive for the regeneration of the myelin sheath. Given the general view that remyelination is, even though limited in human, a natural response to demyelination, targeting pathologically 'dysregulated' extracellular cues and their downstream pathways is regarded as a promising approach toward the enhancement of remyelination by endogenous (or if necessary transplanted) OLG progenitor cells. In this review, we will introduce the extracellular cues that have been implicated in the modulation of (re)myelination. These cues can be soluble, part of the extracellular matrix (ECM) or mediators of cell-cell interactions. Their inhibitory and permissive/promotional roles with regard to remyelination as well as their potential for therapeutic intervention will be discussed.
Collapse
Affiliation(s)
- Natalie A Wheeler
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States.
| |
Collapse
|
12
|
Comabella M, Cantó E, Nurtdinov R, Río J, Villar LM, Picón C, Castilló J, Fissolo N, Aymerich X, Auger C, Rovira A, Montalban X. MRI phenotypes with high neurodegeneration are associated with peripheral blood B-cell changes. Hum Mol Genet 2015; 25:308-16. [DOI: 10.1093/hmg/ddv473] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/15/2015] [Indexed: 11/13/2022] Open
|
13
|
D'Ambrosio A, Pontecorvo S, Colasanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev 2015; 14:1097-110. [PMID: 26226413 DOI: 10.1016/j.autrev.2015.07.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis is the most common autoimmune disorder affecting the central nervous system. The heterogeneity of pathophysiological processes in MS contributes to the highly variable course of the disease and unpredictable response to therapies. The major focus of the research on MS is the identification of biomarkers in biological fluids, such as cerebrospinal fluid or blood, to guide patient management reliably. Because of the difficulties in obtaining spinal fluid samples and the necessity for lumbar puncture to make a diagnosis has reduced, the research of blood-based biomarkers may provide increasingly important tools for clinical practice. However, currently there are no clearly established MS blood-based biomarkers. The availability of reliable biomarkers could radically alter the management of MS at critical phases of the disease spectrum, allowing for intervention strategies that may prevent evolution to long-term neurological disability. This article provides an overview of this research field and focuses on recent advances in blood-based biomarker research.
Collapse
Affiliation(s)
- Antonella D'Ambrosio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Pontecorvo
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Tania Colasanti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Zamboni
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Ada Francia
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Paola Margutti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
14
|
Raphael I, Webb J, Stuve O, Haskins W, Forsthuber T. Body fluid biomarkers in multiple sclerosis: how far we have come and how they could affect the clinic now and in the future. Expert Rev Clin Immunol 2014; 11:69-91. [PMID: 25523168 DOI: 10.1586/1744666x.2015.991315] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system, which affects over 2.5 million people worldwide. Although MS has been extensively studied, many challenges still remain in regards to treatment, diagnosis and prognosis. Typically, prognosis and individual responses to treatment are evaluated by clinical tests such as the expanded disability status scale, MRI and presence of oligoclonal bands in the cerebrospinal fluid. However, none of these measures correlates strongly with treatment efficacy or disease progression across heterogeneous patient populations and subtypes of MS. Numerous studies over the past decades have attempted to identify sensitive and specific biomarkers for diagnosis, prognosis and treatment efficacy of MS. The objective of this article is to review and discuss the current literature on body fluid biomarkers in MS, including research on potential biomarker candidates in the areas of miRNA, mRNA, lipids and proteins.
Collapse
Affiliation(s)
- Itay Raphael
- University of Texas San Antonio - Biology, San Antonio, TX, USA
| | | | | | | | | |
Collapse
|
15
|
Jonas A, Thiem S, Kuhlmann T, Wagener R, Aszodi A, Nowell C, Hagemeier K, Laverick L, Perreau V, Jokubaitis V, Emery B, Kilpatrick T, Butzkueven H, Gresle M. Axonally derived matrilin-2 induces proinflammatory responses that exacerbate autoimmune neuroinflammation. J Clin Invest 2014; 124:5042-56. [PMID: 25329699 DOI: 10.1172/jci71385] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/11/2014] [Indexed: 01/14/2023] Open
Abstract
In patients with multiple sclerosis (MS) and mice with experimental autoimmune encephalomyelitis (EAE), inflammatory axonal injury is a major determinant of disability; however, the drivers of this injury are incompletely understood. Here, we used the EAE model and determined that the extracellular matrix protein matrilin-2 (MATN2) is an endogenous neuronal molecule that is regulated in association with inflammatory axonal injury. Compared with WT mice, mice harboring a deletion of Matn2 exhibited reduced disease severity and axon damage following induction of EAE. Evaluation of neuron-macrophage cocultures revealed that exogenous MATN2 specifically signals through TLR4 and directly induces expression of proinflammatory genes in macrophages, promoting axonal damage. Moreover, the MATN2-induced proinflammatory response was attenuated greatly in macrophages from Myd88 KO mice. Examination of brain sections from patients with MS revealed that MATN2 is expressed in lesions but not in normal-appearing white matter. Together, our results indicate that MATN2 is a deleterious endogenous neuroaxonal injury response signal that activates innate immune cells and could contribute to early axonal damage in CNS inflammatory diseases like MS.
Collapse
|
16
|
Tao Y, Zhang X, Chopra M, Kim MJ, Buch KR, Kong D, Jin J, Tang Y, Zhu H, Jewells V, Markovic-Plese S. The role of endogenous IFN-β in the regulation of Th17 responses in patients with relapsing-remitting multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2014; 192:5610-7. [PMID: 24850724 DOI: 10.4049/jimmunol.1302580] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IFN-β has been used as a first-line therapy for relapsing-remitting multiple sclerosis (RRMS). Because only a few studies have addressed the role of endogenous IFN-β in the pathogenesis of the disease, our objective was to characterize its role in the transcriptional regulation of pathogenic Th17 cytokines in patients with RRMS. In vitro studies have demonstrated that IFN-β inhibits IL-17A, IL-17F, IL-21, IL-22, and IFN-γ secretion in CD4(+) lymphocytes through the induction of suppressor of cytokine secretion 1 and suppressor of cytokine secretion 3. We found that patients with RRMS have increased serum and cerebrospinal fluid Th17 (IL-17A and IL-17F) cytokine levels in comparison with the control subjects, suggesting that deficient endogenous IFN-β secretion or signaling can contribute to the dysregulation of those pathogenic cytokines in CD4(+) cells. We identified that the endogenous IFN-β from serum of RRMS patients induced a significantly lower IFN-inducible gene expression in comparison with healthy controls. In addition, in vitro studies have revealed deficient endogenous and exogenous IFN-β signaling in the CD4(+) cells derived from patients with MS. Interestingly, upon inhibition of the endogenous IFN-β signaling by silencing IFN regulatory factor (IRF) 7 gene expression, the resting CD4(+) T cells secreted significantly higher level of IL-17A, IL-17F, IL-21, IL-22, and IL-9, suggesting that endogenous IFN-β suppresses the secretion of these pathogenic cytokines. In vivo recombinant IFN-β-1a treatment induced IFNAR1 and its downstream signaling molecules' gene expression, suggesting that treatment reconstitutes a deficient endogenous IFN-β regulation of the CD4(+) T cells' pathogenic cytokine production in patients with MS.
Collapse
Affiliation(s)
- Yazhong Tao
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Xin Zhang
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Manisha Chopra
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Ming-Jeong Kim
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Kinnari R Buch
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dehan Kong
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jianping Jin
- Center for Bioinformatics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yunan Tang
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Valerie Jewells
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Silva Markovic-Plese
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
17
|
Raddatz BBR, Hansmann F, Spitzbarth I, Kalkuhl A, Deschl U, Baumgärtner W, Ulrich R. Transcriptomic meta-analysis of multiple sclerosis and its experimental models. PLoS One 2014; 9:e86643. [PMID: 24475162 PMCID: PMC3903571 DOI: 10.1371/journal.pone.0086643] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/12/2013] [Indexed: 12/19/2022] Open
Abstract
Background Multiple microarray analyses of multiple sclerosis (MS) and its experimental models have been published in the last years. Objective Meta-analyses integrate the information from multiple studies and are suggested to be a powerful approach in detecting highly relevant and commonly affected pathways. Data sources ArrayExpress, Gene Expression Omnibus and PubMed databases were screened for microarray gene expression profiling studies of MS and its experimental animal models. Study eligibility criteria Studies comparing central nervous system (CNS) samples of diseased versus healthy individuals with n >1 per group and publically available raw data were selected. Material and Methods Included conditions for re-analysis of differentially expressed genes (DEGs) were MS, myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) in rats, proteolipid protein-induced EAE in mice, Theiler’s murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), and a transgenic tumor necrosis factor-overexpressing mouse model (TNFtg). Since solely a single MS raw data set fulfilled the inclusion criteria, a merged list containing the DEGs from two MS-studies was additionally included. Cross-study analysis was performed employing list comparisons of DEGs and alternatively Gene Set Enrichment Analysis (GSEA). Results The intersection of DEGs in MS, EAE, TMEV-IDD, and TNFtg contained 12 genes related to macrophage functions. The intersection of EAE, TMEV-IDD and TNFtg comprised 40 DEGs, functionally related to positive regulation of immune response. Over and above, GSEA identified substantially more differentially regulated pathways including coagulation and JAK/STAT-signaling. Conclusion A meta-analysis based on a simple comparison of DEGs is over-conservative. In contrast, the more experimental GSEA approach identified both, a priori anticipated as well as promising new candidate pathways.
Collapse
Affiliation(s)
- Barbara B. R. Raddatz
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ingo Spitzbarth
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Arno Kalkuhl
- Department of Non-Clinical Drug Safety, Boehringer Ingelheim Pharma GmbH&Co KG, Biberach (Riß), Germany
| | - Ulrich Deschl
- Department of Non-Clinical Drug Safety, Boehringer Ingelheim Pharma GmbH&Co KG, Biberach (Riß), Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Reiner Ulrich
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
18
|
Abstract
Familial aggregation and the studies of twins indicate that heredity contributes to multiple sclerosis (MS) risk. Immunologic studies of leukocyte antigens subsequently followed by gene-mapping techniques identified the primary MS susceptibility locus to be within the major histocompatibility complex (MHC). The primary risk allele is HLA-DRB1*15, although other alleles of this gene also influence MS susceptibility. Other genes within the MHC also contribute to MS susceptibility. Genome-wide association studies have identified over 50 additional common variants of genes across the genome. Estimates suggest that there may be as many as 200 genes involved in MS susceptibility. In addition to these common polymorphisms, studies have identified several rare risk alleles in some families. Interestingly, the majority of the genes identified have known immunologic functions and many contribute to the risk of inheriting other autoimmune diseases. Genetic variants in the vitamin D metabolic pathway have also been identified. That vitamin D contributes to MS susceptibility as both an environmental as well as genetic risk factor underscores the importance of this metabolic pathway in disease pathogenesis. Current efforts are focused on understanding how the myriad of genetic risk alleles interact within networks to influence MS risk at family level as well as within populations.
Collapse
Affiliation(s)
- Bruce A C Cree
- Department of Neurology, University of California, San Francisco, USA.
| |
Collapse
|
19
|
|
20
|
Abstract
Multiple sclerosis (MS) patients are classified as either having relapsing onset or progressive onset disease, also known as primary progressive MS (PPMS). Relative to relapsing onset patients, PPMS patients are older at disease onset, are equally likely to be men or women, and have more rapid accumulation of disability that does not respond well to treatments used in relapsing onset MS. Although estimates vary, 5-15% of all MS patients have a PPMS disease course. Genetic variance is a proposed determinant of MS disease course. If distinct genes associated with PPMS were identified study of these genes might lead to an understanding of the biology underlying disease progression and neural degeneration that are the hallmarks of PPMS. These genes and their biological pathways might also represent therapeutic targets. This chapter systematically reviews the PPMS genetic literature. Despite the intuitively appealing notion that differences between PPMS and relapsing onset MS are due to genetics, definite differences associated with these phenotypes at the major histocompatibility complex or elsewhere in the genome have not been found. Recent large-scale genome wide screens identified multiple genes associated with MS susceptibility outside the MHC. The genetic variants identified thus far make only weak individual contributions to MS susceptibility. If the genetic effects that contribute to the differences between PPMS and relapsing MS are similar in magnitude to those that distinguish MS from healthy controls then, given the relative scarcity of the PPMS phenotype, very large datasets will be needed to identify PPMS associated genes. International collaborative efforts could provide the means to identify such genes. Alternately, it is possible that factors other than genetics underlie the differences between these clinical phenotypes.
Collapse
Affiliation(s)
- Bruce A C Cree
- Department of Neurology, University of California, San Francisco, USA.
| |
Collapse
|
21
|
Mansilla MJ, Comabella M, Río J, Castilló J, Castillo M, Martin R, Montalban X, Espejo C. Up-regulation of inducible heat shock protein-70 expression in multiple sclerosis patients. Autoimmunity 2013; 47:127-33. [PMID: 24328534 DOI: 10.3109/08916934.2013.866104] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inducible heat shock protein (HSP)70 (HSP70-1A and HSP70-1B proteins) is a chaperone responsible for assisting proper protein folding. Following stress conditions, HSP70 is highly up-regulated to mediate cytoprotective functions. In addition, HSP70 is able to trigger innate and adaptive immune responses that promote the immune recognition of antigens and to act as a cytokine when it is released. The data in the literature are controversial with regard to expression studies in peripheral blood mononuclear cells (PBMCs). In the present study, we aimed to examine if alterations of HSP70-1A/B expression are involved in the autoimmune pathogenesis of multiple sclerosis (MS). We determined both mRNA and protein expression in PBMCs of MS patients and healthy donors (HDs). We found a baseline increased expression of the HSPA1A gene in PBMCs from MS patients compared with HDs. Gene expression findings were associated with an increased protein expression of HSP70-1A/B in T lymphocytes (CD4+ and CD8+) and monocytes from MS patients under basal conditions that may reflect the immunological activation occurring in MS patients. We also provided evidence that heat shock (HS) stimulus induced HSP70-1A/B protein expression in HDs and MS patients, and that HS-induced HSP70-1A/B protein expression in monocytes correlated with the number of T2 lesions at baseline in MS patients. However, after lipopolysaccharide inflammatory stimulus, monocytes from MS patients failed to induce HSP70-1A/B protein expression. Our data hint at altered immune responses in MS and may indicate either a state of chronic stress or increased vulnerability to physiological immune responses in MS patients.
Collapse
Affiliation(s)
- María José Mansilla
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (CEM-Cat), Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona , Barcelona , Spain and
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Sormani MP, De Stefano N. Defining and scoring response to IFN-β in multiple sclerosis. Nat Rev Neurol 2013; 9:504-12. [PMID: 23897407 DOI: 10.1038/nrneurol.2013.146] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The advent of a large number of new therapies for multiple sclerosis (MS) warrants the development of tools that enable selection of the best treatment option for each new patient with MS. Evidence from clinical trials clearly supports the efficacy of IFN-β for the treatment of MS, but few factors that predict a response to this drug in individual patients have emerged. This deficit might be due, at least in part, to the lack of a standardized definition of the clinical outcomes that signify improvement or worsening of the disease. MRI markers and clinical relapses have been the most widely studied short-term factors to predict long-term response to IFN-β, although the results are conflicting. Recently, integrated strategies combining MRI and clinical markers in scoring systems have provided a potentially useful approach for the management of patients with MS. In this Review, we focus on the many definitions of clinical response to IFN-β and explore the markers that can be used to predict this response. We also highlight advantages and limitations of the existing scoring systems in light of future expansion of these models to biological markers and to other classes of emerging therapies for MS.
Collapse
Affiliation(s)
- Maria Pia Sormani
- Department of Health Sciences (DISSAL), University of Genoa, Via Pastore 1, Genoa 16132, Italy. mariapia.sormani@ unige.it
| | | |
Collapse
|
23
|
Apperson ML, Tian Y, Stamova B, Ander BP, Jickling GC, Agius MA, Sharp FR. Genome wide differences of gene expression associated with HLA-DRB1 genotype in multiple sclerosis: a pilot study. J Neuroimmunol 2013; 257:90-6. [PMID: 23477965 DOI: 10.1016/j.jneuroim.2013.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 01/21/2023]
Abstract
Using two microarray platforms, we identify HLA-DRB5 as the most highly expressed gene in MS compared to healthy subjects. As expected, HLA-DRB5 expression was associated with the HLA-DRB1*1501 MS susceptibility allele. Besides HLA-DRB5, there were 1219 differentially expressed exons (p<0.01, |fold change (FC)|>1.2) that differed between HLA-DRB1*1501 Positive multiple sclerosis subjects (MSP) compared to HLA-DRB1*1501 negative multiple sclerosis subjects (MSN). Analysis of the regulated genes revealed significantly different immune signaling pathways including IL-4 and IL-17 in these two MS genotypes. Different risk alleles appear to be associated with different patterns of gene expression that may reflect differences in pathophysiology of these two MS subtypes. These preliminary data will need to be confirmed in future studies.
Collapse
Affiliation(s)
- Michelle L Apperson
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Almeida A. Genetic determinants of neuronal vulnerability to apoptosis. Cell Mol Life Sci 2013; 70:71-88. [PMID: 22695677 PMCID: PMC11113535 DOI: 10.1007/s00018-012-1029-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 04/22/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022]
Abstract
Apoptosis is a common mode of cell death that contributes to neuronal loss associated with neurodegeneration. Single-nucleotide polymorphisms (SNPs) in chromosomal DNA are contributing factors dictating natural susceptibility of humans to disease. Here, the most common SNPs affecting neuronal vulnerability to apoptosis are reviewed in the context of neurological disorders. Polymorphic variants in genes encoding apoptotic proteins, either from the extrinsic (FAS, TNF-α, CASP8) or the intrinsic (BAX, BCL2, CASP3, CASP9) pathways could be highly valuable in the diagnosis of neurodegenerative diseases and stroke. Interestingly, the Arg72Pro SNP in TP53, the gene encoding tumor suppressor p53, was recently revealed a biomarker of poor prognosis in stroke due to its ability to modulate neuronal apoptotic death. Search for new SNPs responsible for genetic variability to apoptosis will ensure the implementation of novel diagnostic and prognostic tools, as well as therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Angeles Almeida
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
25
|
Sánchez-Pla A, Reverter F, Ruíz de Villa MC, Comabella M. Transcriptomics: mRNA and alternative splicing. J Neuroimmunol 2012; 248:23-31. [PMID: 22626445 DOI: 10.1016/j.jneuroim.2012.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/14/2012] [Accepted: 04/18/2012] [Indexed: 11/27/2022]
Abstract
Transcriptomics has emerged as a powerful approach for biomarker discovery. In the present review, the two main types of high throughput transcriptomic technologies - microarrays and next generation sequencing - that can be used to identify candidate biomarkers are briefly described. Microarrays, the mainstream technology of the last decade, have provided hundreds of valuable datasets in a wide variety of diseases including multiple sclerosis (MS), in which this approach has been used to disentangle different aspects of its complex pathogenesis. RNA-seq, the current next generation sequencing approach, is expected to provide similar power as microarrays but extending their capabilities to aspects up to now more difficult to analyse such as alternative splicing and discovery of novel transcripts.
Collapse
|
26
|
Menon R, Di Dario M, Cordiglieri C, Musio S, La Mantia L, Milanese C, Di Stefano AL, Crabbio M, Franciotta D, Bergamaschi R, Pedotti R, Medico E, Farina C. Gender-based blood transcriptomes and interactomes in multiple sclerosis: Involvement of SP1 dependent gene transcription. J Autoimmun 2012; 38:J144-55. [DOI: 10.1016/j.jaut.2011.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/11/2011] [Accepted: 11/12/2011] [Indexed: 12/22/2022]
|
27
|
Arevalo-Martin A, Molina-Holgado E, Guaza C. A CB₁/CB₂ receptor agonist, WIN 55,212-2, exerts its therapeutic effect in a viral autoimmune model of multiple sclerosis by restoring self-tolerance to myelin. Neuropharmacology 2012; 63:385-93. [PMID: 22561283 DOI: 10.1016/j.neuropharm.2012.04.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/19/2012] [Accepted: 04/17/2012] [Indexed: 01/05/2023]
Abstract
Infection of mice with Theiler's murine encephalomyelitis virus (TMEV) leads to the development of TMEV-induced demyelinating disease (TMEV-IDD), an autoimmune, demyelinating and neurodegenerative pathology that serves as a model of multiple sclerosis. Activation of endogenous CB₁/CB₂ cannabinoid receptors inhibits inflammation and improves the clinical status of TMEV-IDD animals. In the present study, mice with established TMEV-IDD were treated with the CB₁/CB₂ receptor agonist WIN 55,212-2 (WIN), which restored self-tolerance to a myelin self-antigen while ameliorating the disease in a long-term manner. Accordingly, disruption of self-tolerance with cyclophosphamide provoked chronic relapse. Furthermore, transfer of splenocytes from WIN-treated TMEV-IDD mice to TMEV-infected mice at disease onset prevented the autoimmune inflammatory response and motor impairment. The therapeutic effect of WIN correlated with a decrease in the activation of CD4⁺CD25⁺Foxp3⁻ T cells and an increase in regulatory CD4⁺CD25⁺Foxp3⁺ T cells in the CNS, along with alterations in the cytokine and chemokine milieu. These findings demonstrate for the first time that the suppression of autoimmune responses to myelin antigens underlies the therapeutic effect of CB₁/CB₂ cannabinoid agonists in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Angel Arevalo-Martin
- Laboratorio de Neuroinflamación, Hospital Nacional de Paraplejicos, SESCAM, Finca la Peraleda s/n., Toledo 45071, Spain.
| | | | | |
Collapse
|
28
|
Dutta R, Trapp BD. Gene expression profiling in multiple sclerosis brain. Neurobiol Dis 2012; 45:108-14. [PMID: 21147224 PMCID: PMC3066282 DOI: 10.1016/j.nbd.2010.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/29/2010] [Accepted: 12/02/2010] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system and the leading cause of non-traumatic neurological disability in young adults in the United States and Europe. The clinical disease course is variable and starts with reversible episodes of neurological disability in the third or fourth decade of life. Microarray-based comparative gene profiling provides a snapshot of genes underlying a particular condition. Several large scale microarray studies have been conducted using brain tissue from MS patients. In this review, we summarize existing data from different gene expression profiling studies and how they relate to understand the pathogenesis of MS.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
29
|
Abstract
Personalized medicine is a broad and rapidly advancing field of health care that is informed by each person's unique clinical, genetic, genomic, and environmental information. Personalized medicine depends on multidisciplinary health care teams and integrated technologies (e.g., clinical decision support) to utilize our molecular understanding of disease in order to optimize preventive health care strategies. Human genome information now allows providers to create optimized care plans at every stage of a disease, shifting the focus from reactive to preventive health care. The further integration of personalized medicine into the clinical workflow requires overcoming several barriers in education, accessibility, regulation, and reimbursement. This review focuses on providing a comprehensive understanding of personalized medicine, from scientific discovery at the laboratory bench to integration of these novel ways of understanding human biology at the bedside.
Collapse
Affiliation(s)
- Isaac S Chan
- Center for Genomic Medicine, Institute for Genome Sciences & Policy, Duke University, Durham, North Carolina 27708, USA
| | | |
Collapse
|
30
|
Abstract
A thorough understanding of the structure and biology of a biotherapeutic is crucial to defining a suitable strategy for pharmacokinetic characterization in proof-of-concept disease models, toxicology species as well as the healthy and disease indication patient populations. This manuscript summarizes parameters that impact bioanalytical strategy for over 50 biotherapeutics indicated for the treatment of oncology, rheumatoid arthritis, allergy, multiple sclerosis, hematology, metabolism and infectious disease. We have addressed numerous therapeutic modalities including chimeric, humanized and fully human monoclonal antibodies, replacement proteins, peptides and fusion proteins, including polyethylene glycol and Fc fusions, as well as antibody–drug conjugates. With the rapid evolution of biotherapeutics over the last 20 years and the contraction of the pharmaceutical and biotechnology labor force, efficient workflow management becomes a crucial bioanalytical component. Thus, we have also addressed new technologies that have demonstrated either increased throughput or enhanced characterization, including Meso Scale Discovery, Gyrolab and affinity MS.
Collapse
|
31
|
Quest for new genomic and proteomic biomarkers in neurology. Transl Neurosci 2011. [DOI: 10.2478/s13380-011-0005-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe possibility of identifying novel biomarkers for neurodegenerative diseases has been greatly enhanced with recent advances in genomics and proteomics. Novel technologies have the potential to hasten the development of new biomarkers useful as predictors of disease etiology and outcome, as well as responsiveness to therapy. Disease-modifying new therapies are very much needed in modern approaches to treatment of neurodegenerative diseases. Current progress in the field encounters a degree of skepticism about the reliability of genomic and proteomic data and its relevance for clinical applications. Standard operating procedures covering sample collection, methodology and statistical analysis need to be fully developed and strictly adhered to in order to assure reproducible and clinically relevant results. Previous studies involving patients with neurodegenerative diseases show promise in using genomic and proteomic approaches for development of new biomarkers. Confirmation of any novel biomarker in multiple independent patient cohorts and correlation of the improvement in biomarker endpoint with clinical improvement in longitudinal patient studies remains crucial for future successful application. We propose that a combination of approaches in biomarker discovery may in the end lead to identification of promising candidates at DNA, RNA, protein and small molecule level.
Collapse
|
32
|
Mix E, Meyer-Rienecker H, Hartung HP, Zettl UK. Animal models of multiple sclerosis--potentials and limitations. Prog Neurobiol 2010; 92:386-404. [PMID: 20558237 PMCID: PMC7117060 DOI: 10.1016/j.pneurobio.2010.06.005] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 06/01/2010] [Accepted: 06/07/2010] [Indexed: 12/17/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is still the most widely accepted animal model of multiple sclerosis (MS). Different types of EAE have been developed in order to investigate pathogenetic, clinical and therapeutic aspects of the heterogenic human disease. Generally, investigations in EAE are more suitable for the analysis of immunogenetic elements (major histocompatibility complex restriction and candidate risk genes) and for the study of histopathological features (inflammation, demyelination and degeneration) of the disease than for screening of new treatments. Recent studies in new EAE models, especially in transgenic ones, have in connection with new analytical techniques such as microarray assays provided a deeper insight into the pathogenic cellular and molecular mechanisms of EAE and potentially of MS. For example, it was possible to better delineate the role of soluble pro-inflammatory (tumor necrosis factor-α, interferon-γ and interleukins 1, 12 and 23), anti-inflammatory (transforming growth factor-β and interleukins 4, 10, 27 and 35) and neurotrophic factors (ciliary neurotrophic factor and brain-derived neurotrophic factor). Also, the regulatory and effector functions of distinct immune cell subpopulations such as CD4+ Th1, Th2, Th3 and Th17 cells, CD4+FoxP3+ Treg cells, CD8+ Tc1 and Tc2, B cells and γδ+ T cells have been disclosed in more detail. The new insights may help to identify novel targets for the treatment of MS. However, translation of the experimental results into the clinical practice requires prudence and great caution.
Collapse
Key Words
- apc, antigen-presenting cell
- at-eae, adoptive transfer eae
- bbb, blood–brain barrier
- bdnf, brain-derived neurotrophic factor
- cd, cluster of differentiation
- cns, central nervous system
- cntf, ciliary neurotrophic factor
- eae, experimental autoimmune encephalomyelitis
- hla, human leukocyte antigen
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ivig, intravenous immunoglobulin
- mab, monoclonal antibody
- mbp, myelin basic protein
- mhc, major histocompatibility complex
- mog, myelin oligodendrocyte glycoprotein
- mp, methylprednisolone
- mri, magnetic resonance imaging
- ms, multiple sclerosis
- nk, natural killer
- odc, oligodendrocyte
- qtl, quantitative trait locus
- plp, proteolipid protein
- tc, cytotoxic t cell
- tcr, t cell receptor
- tgf, transforming growth factor
- th cell, helper t cell
- tnf, tumor necrosis factor
- animal model
- autoimmunity
- experimental autoimmune encephalomyelitis
- immunogenetics
- immunomodulatory therapy
- multiple sclerosis
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Clinical Trials as Topic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Microarray Analysis
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/physiopathology
- Multiple Sclerosis/therapy
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Germany
| | | | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-University, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, Germany
| |
Collapse
|
33
|
Hecker M, Goertsches RH, Fatum C, Koczan D, Thiesen HJ, Guthke R, Zettl UK. Network analysis of transcriptional regulation in response to intramuscular interferon-β-1a multiple sclerosis treatment. THE PHARMACOGENOMICS JOURNAL 2010; 12:134-46. [PMID: 20956993 DOI: 10.1038/tpj.2010.77] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interferon-β (IFN-β) is one of the major drugs for multiple sclerosis (MS) treatment. The purpose of this study was to characterize the transcriptional effects induced by intramuscular IFN-β-1a therapy in patients with relapsing-remitting form of MS. By using Affymetrix DNA microarrays, we obtained genome-wide expression profiles of peripheral blood mononuclear cells of 24 MS patients within the first 4 weeks of IFN-β administration. We identified 121 genes that were significantly up- or downregulated compared with baseline, with stronger changed expression at 1 week after start of therapy. Eleven transcription factor-binding sites (TFBS) are overrepresented in the regulatory regions of these genes, including those of IFN regulatory factors and NF-κB. We then applied TFBS-integrating least angle regression, a novel integrative algorithm for deriving gene regulatory networks from gene expression data and TFBS information, to reconstruct the underlying network of molecular interactions. An NF-κB-centered sub-network of genes was highly expressed in patients with IFN-β-related side effects. Expression alterations were confirmed by real-time PCR and literature mining was applied to evaluate network inference accuracy.
Collapse
Affiliation(s)
- M Hecker
- Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Jena, Germany.
| | | | | | | | | | | | | |
Collapse
|
34
|
Overlapping and distinct mechanisms of action of multiple sclerosis therapies. Clin Neurol Neurosurg 2010; 112:583-91. [DOI: 10.1016/j.clineuro.2010.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 04/27/2010] [Accepted: 05/04/2010] [Indexed: 11/18/2022]
|
35
|
Mechelli R, Annibali V, Ristori G, Vittori D, Coarelli G, Salvetti M. Multiple sclerosis etiology: beyond genes and environment. Expert Rev Clin Immunol 2010; 6:481-90. [PMID: 20441432 DOI: 10.1586/eci.10.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a disorder of the CNS with inflammatory and neurodegenerative components. The etiology is unknown, but there is evidence for a role of both genetic and environmental factors. Among the heritable factors, MHC class II genes are strongly involved, as well as genes coding for others molecules of immunological relevance, genes controlling neurobiological pathways and genes of unknown function. Among nonheritable factors, many infectious agents (mainly viruses) and environmental factors (e.g., smoke, sun exposition and diet) seem to be of etiologic importance. Here, we report and discuss recent findings in MS on largely unexplored fields: the alternative splicing of mRNAs and regulatory noncoding RNAs, the major sources of transcriptome diversity; and epigenetic changes with special attention paid to DNA methylation and histone acetylation, the main regulators of gene expression.
Collapse
Affiliation(s)
- Rosella Mechelli
- Neurology and Center for Experimental Neurological Therapies, S. Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | | | | | | | | | | |
Collapse
|
36
|
Gäckler A, Eickelmann AK, Brors D, Dazert S, Epplen JT, Kunstmann E. Positive family history of idiopathic sudden sensorineural hearing loss. Eur Arch Otorhinolaryngol 2010; 267:1843-8. [PMID: 20593290 DOI: 10.1007/s00405-010-1310-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Accepted: 06/14/2010] [Indexed: 11/24/2022]
Abstract
Idiopathic sudden sensorineural hearing loss (ISSNHL) is a heterogenic disease. Multiple factors influencing aetiology and prognosis are discussed. A retrospective clinical characterisation and analysis of family history of ISSNHL patients was performed to investigate influences on the disease. 186 inpatients diagnosed with ISSNHL were characterised by health records and a standardised questionnaire. Audiograms were observed. 75 controls that had never experienced an event of ISSNHL were questioned about family members being affected by ISSNHL. 63.4% of all patients could be assigned to at least one group with similar causes of ISSNHL (noise exposure, positive family history, infectious diseases, hypothyroidism and fibromuscular dysplasia). A positive family history for ISSNHL has not been reported so far. Therefore, we accentuated the characterisation of patients with positive family history. 21.4% affirmed a positive family history. In ten families, at least two family members were reported as ISSNHL patients. In comparison with patients with negative family history, they tend to be younger, experience more events of ISSNHL and show less improvement of hearing abilities under therapeutic treatment (non-significant). Differences intensified between smokers with positive family history and non-smokers with negative family history. Differences concerning average age were statistically significant (p = 0.001). Within 75 controls 11 families were reported with one member being affected by ISSNHL. In the control group we did not detect any family with more than one ISSNHL patient. The results indicated that patients with positive family history tend to have an aggravated course of ISSNHL. Further studies should help to confirm these results and to identify environmental or genetic factors leading to ISSNHL. This might support a better understanding of the aetiology of ISSNHL and offer new possibilities for prevention and therapy.
Collapse
Affiliation(s)
- A Gäckler
- Department of Human Genetics, Ruhr-University Bochum, Universitätsstrasse 150, 44801, Bochum, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Prinz M, Kalinke U. New lessons about old molecules: how type I interferons shape Th1/Th17-mediated autoimmunity in the CNS. Trends Mol Med 2010; 16:379-86. [PMID: 20591737 DOI: 10.1016/j.molmed.2010.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 01/02/2023]
Abstract
Type I interferons (IFN-alpha and IFN-beta) were discovered more than five decades ago and are widely used for the treatment of human autoimmune diseases such as multiple sclerosis (MS). Despite their highly beneficial features, the precise mechanism of action remains speculative. Given the frequent side effects of IFN-alpha/beta therapy, understanding its action in an in vivo setting is vital to further improve this therapeutic approach. Major advances in our understanding of the IFN biology have recently been made and are particularly based on the combination of powerful genome-wide expression analysis in humans with gene-targeting techniques available for basic research. The recent discovery of a novel T-cell subset, Th17 cells, sheds new light on type I IFNs in MS.
Collapse
Affiliation(s)
- Marco Prinz
- Department of Neuropathology, University of Freiburg, Breisacher Str. 64, D-79106 Freiburg, Germany.
| | | |
Collapse
|
38
|
Genomics in multiple sclerosis. Clin Neurol Neurosurg 2010; 112:621-4. [PMID: 20471158 DOI: 10.1016/j.clineuro.2010.03.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 03/23/2010] [Accepted: 03/29/2010] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis (MS) is chronic, inflammatory disease of the central nervous system that mainly affects young adults and is characterized with dissemination of demyelinating lesions in time and space. It is well known that MS is very heterogeneous disease, so biomarkers that would reliably determine disease course, outcome or treatment response in early stages of the disease (preferentially clinically isolated syndrome) are desperately needed. Genome-wide expression analysis represents the profile of all genes in a certain tissue or cell population in a certain time point. Therefore, as the sequence of the human genome is entirely known, it is possible to analyze any given human gene in any given context. This review will discuss results and possible applications of genome-wide expression studies in brain tissue and blood samples of MS patients.
Collapse
|
39
|
Tseveleki V, Rubio R, Vamvakas SS, White J, Taoufik E, Petit E, Quackenbush J, Probert L. Comparative gene expression analysis in mouse models for multiple sclerosis, Alzheimer's disease and stroke for identifying commonly regulated and disease-specific gene changes. Genomics 2010; 96:82-91. [PMID: 20435134 DOI: 10.1016/j.ygeno.2010.04.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/22/2010] [Accepted: 04/22/2010] [Indexed: 12/17/2022]
Abstract
The brain responds to injury and infection by activating innate defense and tissue repair mechanisms. Working upon the hypothesis that the brain defense response involves common genes and pathways across diverse pathologies, we analysed global gene expression in brain from mouse models representing three major central nervous system disorders, cerebral stroke, multiple sclerosis and Alzheimer's disease compared to normal brain using DNA microarray expression profiling. A comparison of dysregulated genes across disease models revealed common genes and pathways including key components of estrogen and TGF-beta signaling pathways that have been associated with neuroprotection as well as a neurodegeneration mediator, TRPM7. Further, for each disease model, we discovered collections of differentially expressed genes that provide novel insight into the individual pathology and its associated mechanisms. Our data provide a resource for exploring the complex molecular mechanisms that underlie brain neurodegeneration and a new approach for identifying generic and disease-specific targets for therapy.
Collapse
|
40
|
deLuca LES, Pikor NB, O'Leary J, Galicia-Rosas G, Ward LA, Defreitas D, Finlay TM, Ousman SS, Osborne LR, Gommerman JL. Substrain differences reveal novel disease-modifying gene candidates that alter the clinical course of a rodent model of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2010; 184:3174-85. [PMID: 20173032 DOI: 10.4049/jimmunol.0902881] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a rodent model of multiple sclerosis that is executed in animals by immunization with myelin Ag in adjuvant. The SJL/J autoimmune-prone strain of mouse has been used to model relapsing-remitting multiple sclerosis. However, significant variations in peak scores, timing of onset, and incidence are observed among laboratories, with the postacute (relapse) phase of the disease exhibiting significant inconsistency. We characterized two substrains of SJL/J mice that exhibit profoundly different EAE disease parameters. Induction of EAE in the first SJL/J substrain resulted in many cases of chronic EAE that was dominated by an aggressive B cell response to the immunizing Ag and to endogenous CNS Ags. In contrast, the other SJL/J substrain exhibited a relapsing-remitting form of EAE concomitant with an elevated number of cytokine-producing CD4(+) T cells in the CNS. Exploiting these interstrain differences, we performed a genome-wide copy number analysis on the two disparate SJL/J substrains and discovered numerous gene-dosage differences. In particular, one inflammation-associated gene, Naip1, was present at a higher copy number in the SJL/J substrain that exhibited relapsing-remitting EAE. These results demonstrate that substrain differences, perhaps at the level of genomic copy number, can account for variability in the postacute phase of EAE and may drive chronic versus relapsing disease.
Collapse
|
41
|
Linker RA, Brechlin P, Jesse S, Steinacker P, Lee DH, Asif AR, Jahn O, Tumani H, Gold R, Otto M. Proteome profiling in murine models of multiple sclerosis: identification of stage specific markers and culprits for tissue damage. PLoS One 2009; 4:e7624. [PMID: 19865482 PMCID: PMC2765069 DOI: 10.1371/journal.pone.0007624] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2009] [Accepted: 09/10/2009] [Indexed: 12/20/2022] Open
Abstract
The identification of new biomarkers is of high interest for the prediction of the disease course and also for the identification of pathomechanisms in multiple sclerosis (MS). To specify markers of the chronic disease phase, we performed proteome profiling during the later phase of myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE, day 35 after immunization) as a model disease mimicking many aspects of secondary progressive MS. In comparison to healthy controls, high resolution 2 dimensional gel electrophoresis revealed a number of regulated proteins, among them glial fibrilary acidic protein (GFAP). Phase specific up-regulation of GFAP in chronic EAE was confirmed by western blotting and immunohistochemistry. Protein levels of GFAP were also increased in the cerebrospinal fluid of MS patients with specificity for the secondary progressive disease phase. In a next step, proteome profiling of an EAE model with enhanced degenerative mechanisms revealed regulation of alpha-internexin, syntaxin binding protein 1, annexin V and glutamate decarboxylase in the ciliary neurotrophic factor (CNTF) knockout mouse. The identification of these proteins implicate an increased apoptosis and enhanced axonal disintegration and correlate well the described pattern of tissue injury in CNTF −/− mice which involve oligodendrocyte (OL) apoptosis and axonal injury. In summary, our findings underscore the value of proteome analyses as screening method for stage specific biomarkers and for the identification of new culprits for tissue damage in chronic autoimmune demyelination.
Collapse
Affiliation(s)
- Ralf A. Linker
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Peter Brechlin
- Department of Neurodegeneration and Restorative Research, Center of Neurological Medicine, University of Goettingen, Goettingen, Germany
- DFG Research Center for Molecular Physiology of the Brain, Goettingen, Germany
| | - Sarah Jesse
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - D. H. Lee
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Abdul R. Asif
- Department of Clinical Chemistry, University of Goettingen, Goettingen, Germany
| | - Olaf Jahn
- DFG Research Center for Molecular Physiology of the Brain, Goettingen, Germany
- Max-Planck-Institute for Experimental Medicine, Goettingen, Germany
| | | | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
42
|
Río J, Comabella M, Montalban X. Predicting responders to therapies for multiple sclerosis. Nat Rev Neurol 2009; 5:553-60. [DOI: 10.1038/nrneurol.2009.139] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
43
|
Tegla CA, Cudrici C, Rus V, Ito T, Vlaicu S, Singh A, Rus H. Neuroprotective effects of the complement terminal pathway during demyelination: implications for oligodendrocyte survival. J Neuroimmunol 2009; 213:3-11. [PMID: 19577811 DOI: 10.1016/j.jneuroim.2009.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 06/01/2009] [Accepted: 06/04/2009] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system that is mediated by activated lymphocytes, macrophages/microglia, and complement. In MS, the myelin-forming oligodendrocytes (OLGs) are the targets of the immune attack. Experimental evidence indicates that C5b-9 plays a role in demyelination during the acute phase of experimental allergic encephalomyelitis (EAE). Terminal complement C5b-9 complexes are capable of protecting OLGs from apoptosis. During chronic EAE complement C5 promotes axonal preservation, remyelination and provides protection from gliosis. These findings indicate that the activation of complement and C5b-9 assembly can also have protective roles during demyelination.
Collapse
Affiliation(s)
- Cosmin A Tegla
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Zhang X, Jin J, Tang Y, Speer D, Sujkowska D, Markovic-Plese S. IFN-beta1a inhibits the secretion of Th17-polarizing cytokines in human dendritic cells via TLR7 up-regulation. THE JOURNAL OF IMMUNOLOGY 2009; 182:3928-36. [PMID: 19265172 DOI: 10.4049/jimmunol.0802226] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
IFN-beta, an effective therapy against relapsing-remitting multiple sclerosis, is naturally secreted during the innate immune response against viral pathogens. The objective of this study was to characterize the immunomodulatory mechanisms of IFN-beta targeting innate immune response and their effects on dendritic cell (DC)-mediated regulation of T cell differentiation. We found that IFN-beta1a in vitro treatment of human monocyte-derived DCs induced the expression of TLR7 and the members of its downstream signaling pathway, including MyD88, IL-1R-associated kinase 4, and TNF receptor-associated factor 6, while it inhibited the expression of IL-1R. Using small interfering RNA TLR7 gene silencing, we confirmed that IFN-beta1a-induced changes in MyD88, IL-1R-associated kinase 4, and IL-1R expression were dependent on TLR7. TLR7 expression was also necessary for the IFN-beta1a-induced inhibition of IL-1beta and IL-23 and the induction of IL-27 secretion by DCs. Supernatant transfer experiments confirmed that IFN-beta1a-induced changes in DC cytokine secretion inhibit Th17 cell differentiation as evidenced by the inhibition of retinoic acid-related orphan nuclear hormone receptor C and IL-17A gene expression and IL-17A secretion. Our study has identified a novel therapeutic mechanism of IFN-beta1a that selectively targets the autoimmune response in multiple sclerosis.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
45
|
Goertsches RH, Hecker M, Zettl UK. Monitoring of multiple sclerosis immunotherapy: from single candidates to biomarker networks. J Neurol 2009; 255 Suppl 6:48-57. [PMID: 19300960 DOI: 10.1007/s00415-008-6010-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Applying microarray technology to identify new diagnostic and prognostic markers in peripheral blood cells (PBC) after therapeutic intervention opens great perspectives regarding patient subclassification. Three recombinant products of the pleiotropic agent interferon beta (rIFN-beta) are available for disease modifying therapy of relapsing remitting multiple sclerosis (RRMS), a complex inflammatory autoimmune disease that targets the central nervous system. They differ according to formulation, route of administration and dosage regimens. The currently, only partially understood mechanism of action of injected rIFN-beta into human organisms needs provision with accessory key molecules; in addition, the significance of established clinical IFN-beta response criteria that distinguish responding from non-responding patients remain unclear.With respect to these major questions, we discuss promising candidates on the gene transcription level, attained from scientific MS literature that included a longitudinal aspect. Reviewed studies were in part carried out with distinct gene interrogating platforms (GeneArrays; RT-PCR), settings (in vitro; ex vivo), and study designs (drug formulations and regimen; inclusion criteria and clinical endpoints), hampering meaningful meta-analysis. Nevertheless, PBC from therapy-naïve MS patients, rIFN-beta treated MS patients, and healthy controls served to characterize facets of both the disease and its treatment. Hence, the field of MS transcriptomics in immunomodulatory therapy is (by far) not adequately understood and should be embedded into systems biology disciplines, yielding multi-layer analyses that deliver timely identification of MS subjects who will profit from applied rIFN-beta therapy.
Collapse
Affiliation(s)
- Robert H Goertsches
- Department of Neurology, University of Rostock, Gehlsheimer Strasse 20, 18147 Rostock, Germany.
| | | | | |
Collapse
|
46
|
Ulrich R, Kalkuhl A, Deschl U, Baumgärtner W. Machine learning approach identifies new pathways associated with demyelination in a viral model of multiple sclerosis. J Cell Mol Med 2009; 14:434-48. [PMID: 19183246 PMCID: PMC3837619 DOI: 10.1111/j.1582-4934.2008.00646.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Theiler’s murine encephalomyelitis is an experimentally virus-induced inflammatory demyelinating disease of the spinal cord, displaying clinical and pathological similarities to chronic progressive multiple sclerosis. The aim of this study was to identify pathways associated with chronic demyelination using an assumption-free combined microarray and immunohistology approach. Movement control as determined by rotarod assay significantly worsened in Theiler’s murine encephalomyelitis -virus-infected SJL/J mice from 42 to 196 days after infection (dpi). In the spinal cords, inflammatory changes were detected 14 to 196 dpi, and demyelination progressively increased from 42 to 196 dpi. Microarray analysis revealed 1001 differentially expressed genes over the study period. The dominating changes as revealed by k-means and functional annotation clustering included up-regulations related to intrathecal antibody production and antigen processing and presentation via major histocompatibility class II molecules. A random forest machine learning algorithm revealed that down-regulated lipid and cholesterol biosynthesis, differentially expressed neurite morphogenesis and up-regulated toll-like receptor-4-induced pathways were intimately associated with demyelination as measured by immunohistology. Conclusively, although transcriptional changes were dominated by the adaptive immune response, the main pathways associated with demyelination included up-regulation of toll-like receptor 4 and down-regulation of cholesterol biosynthesis. Cholesterol biosynthesis is a rate limiting step of myelination and its down-regulation is suggested to be involved in chronic demyelination by an inhibition of remyelination.
Collapse
Affiliation(s)
- Reiner Ulrich
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg, Hannover, Germany.
| | | | | | | |
Collapse
|
47
|
Montes M, Zhang X, Berthelot L, Laplaud DA, Brouard S, Jin J, Rogan S, Armao D, Jewells V, Soulillou JP, Markovic-Plese S. Oligoclonal myelin-reactive T-cell infiltrates derived from multiple sclerosis lesions are enriched in Th17 cells. Clin Immunol 2008; 130:133-44. [PMID: 18977698 DOI: 10.1016/j.clim.2008.08.030] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/25/2008] [Accepted: 08/29/2008] [Indexed: 12/30/2022]
Abstract
In this study, acute and chronic brain and spinal cord lesions, and normal appearing white matter (NAWM), were resected post-mortem from a patient with aggressive relapsing-remitting multiple sclerosis (MS). T-cell infiltrates from the central nervous system (CNS) lesions and NAWM were separated and characterized in-vitro. All infiltrates showed a proliferative response against multiple myelin peptides. Studies of the T-cell receptor (TCR)Vbeta and Jbeta usage revealed a very skewed repertoire with shared complementarity-determining region (CDR)3 lengths detected in all CNS lesions and NAWM. In the acute lesion, genomic profiling of the infiltrating T-cells revealed up-regulated expression of TCRalpha and beta chain, retinoic acid-related orphan nuclear hormone receptor C (RORC) transcription factor, and multiple cytokine genes that mediate Th17 cell expansion. The differentially expressed genes involved in regulation of Th17 cells represent promising targets for new therapies of relapsing-remitting MS.
Collapse
Affiliation(s)
- Monica Montes
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Weinstock-Guttman B, Bhasi K, Badgett D, Tamaño-Blanco M, Minhas M, Feichter J, Patrick K, Munschauer F, Bakshi R, Ramanathan M. Genomic effects of once-weekly, intramuscular interferon-beta1a treatment after the first dose and on chronic dosing: Relationships to 5-year clinical outcomes in multiple sclerosis patients. J Neuroimmunol 2008; 205:113-25. [PMID: 18950872 DOI: 10.1016/j.jneuroim.2008.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 08/25/2008] [Accepted: 09/02/2008] [Indexed: 01/10/2023]
Abstract
PURPOSE To characterize gene expression in multiple sclerosis (MS) patients after the first dose and chronic dosing of 30 microg, once weekly, intramuscular interferon-beta1a (IFN-beta) and to delineate the pharmacogenomic differences between Good Responders and Partial Responders to IFN-beta therapy. METHODS The treatment responses after the first IFN-beta dose and chronic IFN-beta dosing were assessed in 22 relapsing MS patients (17 females, 5 males; average age: 41.5+/-SD 10.4 years). Gene expression profiles in peripheral blood mononuclear cells were obtained prior to treatment and at 1, 2, 4, 8, 24, 48, 120, 168 h after the first IFN-beta dose and at 1, 6 and 12 months after chronic dosing with once-weekly 30 microg IFN-beta-1a intramuscularly. Repeated measures statistics with false discovery rate control were used. The functional characteristics, biological pathways and transcription factor sites were analyzed. RESULTS Of the 1000 genes modulated following the first dose and upon chronic dosing of IFN-beta in MS patients, approximately 35% were up-regulated and 65% were down- regulated; the percentage of modulated genes in common was approximately 50%. The expression of the pharmacodynamic mRNA markers of IFN-beta effect showed differences in time profiles for the Good Responder and Partial Responders to IFN-beta therapy and the Jak-STAT, TNFRSF10B, IL6, TGFbeta, retinoic acid and CDC42 pathways were differentially modulated. The patients with side effects to therapy showed differences in the TGFbeta1, IFNG/STAT3 and TNF pathways. CONCLUSIONS Gene expression is a valuable tool for understanding the molecular mechanisms of IFN-beta action in MS patients.
Collapse
|
49
|
Oksenberg JR, Baranzini SE, Sawcer S, Hauser SL. The genetics of multiple sclerosis: SNPs to pathways to pathogenesis. Nat Rev Genet 2008; 9:516-26. [PMID: 18542080 DOI: 10.1038/nrg2395] [Citation(s) in RCA: 257] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease and a common cause of neurological disability in young adults. The modest heritability of MS reflects complex genetic effects and multifaceted gene-environment interactions. The human leukocyte antigen (HLA) region is the strongest susceptibility locus for MS, but a genome-wide association study recently identified new susceptibility genes. Progress in high-throughput genotyping and sequencing technologies and a better understanding of the structural organization of the human genome, together with powerful brain-imaging techniques that refine the phenotype, suggest that the tools could finally exist to identify the full set of genes influencing the pathogenesis of MS.
Collapse
Affiliation(s)
- Jorge R Oksenberg
- Department of Neurology, School of Medicine, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, California 94143-0435, USA.
| | | | | | | |
Collapse
|
50
|
Foster CA, Mechtcheriakova D, Storch MK, Balatoni B, Howard LM, Bornancin F, Wlachos A, Sobanov J, Kinnunen A, Baumruker T. FTY720 rescue therapy in the dark agouti rat model of experimental autoimmune encephalomyelitis: expression of central nervous system genes and reversal of blood-brain-barrier damage. Brain Pathol 2008; 19:254-66. [PMID: 18540945 DOI: 10.1111/j.1750-3639.2008.00182.x] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
FTY720 (fingolimod) is an oral sphingosine-1 phosphate (S1P) receptor modulator in phase III development for the treatment of multiple sclerosis. To further investigate its mode of action, we analyzed gene expression in the central nervous system (CNS) during experimental autoimmune encephalomyelitis (EAE). FTY720 downregulated inflammatory genes in addition to vascular adhesion molecules. It decreased the matrix metalloproteinase gene MMP-9 and increased its counterregulator--tissue inhibitor of metalloproteinase, TIMP-1--resulting in a proteolytic balance that favors preservation of blood-brain-barrier (BBB) integrity. Furthermore, FTY720 reduced S1P lyase that increases the S1P concentration in the brain, in line with a marked reversal of neurological deficits and raising the possibility for enhanced triggering of S1P receptors on resident brain cells. This is accompanied by an increase in S1P(1) and S1P(5) in contrast with the attenuation of S1P(3) and S1P(4). Late-stage rescue therapy with FTY720, even up to 1 month after EAE onset, reversed BBB leakiness and reduced demyelination, along with normalization of neurologic function. Our results indicate rapid blockade of ongoing disease processes by FTY720, and structural restoration of the CNS parenchyma, which is likely caused by the inhibition of autoimmune T cell infiltration and direct modulation of microvascular and/or glial cells.
Collapse
Affiliation(s)
- Carolyn A Foster
- Novartis Institutes for BioMedical Research, Brunner Strasse 59, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|