1
|
Myrou A, Barmpagiannos K, Ioakimidou A, Savopoulos C. Molecular Biomarkers in Neurological Diseases: Advances in Diagnosis and Prognosis. Int J Mol Sci 2025; 26:2231. [PMID: 40076852 PMCID: PMC11900390 DOI: 10.3390/ijms26052231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Neurological diseases contribute significantly to disability and mortality, necessitating improved diagnostic and prognostic tools. Advances in molecular biomarkers at genomic, transcriptomic, epigenomic, and proteomic levels have facilitated early disease detection. Notably, neurofilament light chain (NfL) serves as a key biomarker of neurodegeneration, while liquid biopsy techniques enable non-invasive monitoring through exosomal tau, α-synuclein, and inflammatory markers. Artificial intelligence (AI) and multi-omics integration further enhance biomarker discovery, promoting precision medicine. A comprehensive literature review was conducted using PubMed, Scopus, and Web of Science to identify studies (2010-2024) on molecular biomarkers in neurodegenerative and neuroinflammatory disorders. Key findings on genomic mutations, transcriptomic signatures, epigenetic modifications, and protein-based biomarkers were analyzed. The findings highlight the potential of liquid biopsy and multi-omics approaches in improving diagnostic accuracy and therapeutic stratification. Genomic, transcriptomic, and proteomic markers demonstrate utility in early detection and disease monitoring. AI-driven analysis enhances biomarker discovery and clinical application. Despite advancements, challenges remain in biomarker validation, standardization, and clinical implementation. Large-scale longitudinal studies are essential to ensure reliability. AI-powered multi-omics analysis may accelerate biomarker application, ultimately improving patient outcomes in neurological diseases.
Collapse
Affiliation(s)
- Athena Myrou
- Department of Internal Medicine, American Hellenic Educational Progressive Association (AHEPA) University Hospital, 54636 Thessaloniki, Greece; (K.B.)
| | - Konstantinos Barmpagiannos
- Department of Internal Medicine, American Hellenic Educational Progressive Association (AHEPA) University Hospital, 54636 Thessaloniki, Greece; (K.B.)
| | - Aliki Ioakimidou
- Microbiology Laboratory, Department of Immunology, American Hellenic Educational Progressive Association (AHEPA) University Hospital, 54636 Thessaloniki, Greece;
| | - Christos Savopoulos
- Department of Internal Medicine, American Hellenic Educational Progressive Association (AHEPA) University Hospital, 54636 Thessaloniki, Greece; (K.B.)
| |
Collapse
|
2
|
Owens GP, Fellin TJ, Matschulat A, Salas V, Schaller KL, Given KS, Ritchie AM, Navarro A, Blauth K, Hughes EG, Macklin WB, Bennett JL. Pathogenic myelin-specific antibodies in multiple sclerosis target conformational proteolipid protein 1-anchored membrane domains. J Clin Invest 2023; 133:e162731. [PMID: 37561592 PMCID: PMC10541191 DOI: 10.1172/jci162731] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
B cell clonal expansion and cerebrospinal fluid (CSF) oligoclonal IgG bands are established features of the immune response in multiple sclerosis (MS). Clone-specific recombinant monoclonal IgG1 Abs (rAbs) derived from MS patient CSF plasmablasts bound to conformational proteolipid protein 1 (PLP1) membrane complexes and, when injected into mouse brain with human complement, recapitulated histologic features of MS pathology: oligodendrocyte cell loss, complement deposition, and CD68+ phagocyte infiltration. Conformational PLP1 membrane epitopes were complex and governed by the local cholesterol and glycolipid microenvironment. Abs against conformational PLP1 membrane complexes targeted multiple surface epitopes, were enriched within the CSF compartment, and were detected in most MS patients, but not in inflammatory and noninflammatory neurologic controls. CSF PLP1 complex Abs provide a pathogenic autoantibody biomarker specific for MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ethan G. Hughes
- Department of Cell & Developmental Biology
- Program in Neuroscience
| | - Wendy B. Macklin
- Department of Cell & Developmental Biology
- Program in Neuroscience
| | - Jeffrey L. Bennett
- Department of Neurology
- Program in Neuroscience
- Department of Ophthalmology, and
- Program in Immunology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
3
|
Charabati M, Wheeler MA, Weiner HL, Quintana FJ. Multiple sclerosis: Neuroimmune crosstalk and therapeutic targeting. Cell 2023; 186:1309-1327. [PMID: 37001498 PMCID: PMC10119687 DOI: 10.1016/j.cell.2023.03.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/23/2023] [Accepted: 03/03/2023] [Indexed: 04/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system afflicting nearly three million individuals worldwide. Neuroimmune interactions between glial, neural, and immune cells play important roles in MS pathology and offer potential targets for therapeutic intervention. Here, we review underlying risk factors, mechanisms of MS pathogenesis, available disease modifying therapies, and examine the value of emerging technologies, which may address unmet clinical needs and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Marc Charabati
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
4
|
Lanz TV, Robinson WH, Ho PP, Steinman L. Roadmap for understanding mechanisms on how Epstein-Barr virus triggers multiple sclerosis and for translating these discoveries in clinical trials. Clin Transl Immunology 2023; 12:e1438. [PMID: 36815946 PMCID: PMC9933111 DOI: 10.1002/cti2.1438] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Here, we offer a roadmap for what might be studied next in understanding how EBV triggers MS. We focus on two areas: The first area concerns the molecular mechanisms underlying how clonal antibody in the CSF emanates in widespread molecular mimicry to key antigens in the nervous system including GlialCAM, a protein associated with chloride channels. A second and equally high priority in the roadmap concerns various therapeutic approaches that are related to blocking the mechanisms whereby EBV triggers MS. Therapies deserving of attention include clinical trials with antivirals and the development of 'inverse' vaccines based on nucleic acid technologies to control or to eradicate the consequences of EBV infection. High enthusiasm is given to continuation of ongoing clinical trials of cellular adoptive therapy to attack EBV-infected cells. Clinical trials of vaccines to EBV are another area deserving attention. These suggested topics involving research on mechanism, and the design, implementation and performance of well-designed trials are not intended to be an exhaustive list. We have splendid tools available to our community of medical scientists to tackle how EBV triggers MS and then to perhaps change the world with new therapies to potentially eradicate MS, as we have done with nearly complete success for poliomyelitis.
Collapse
|
5
|
Steinman L, Patarca R, Haseltine W. Experimental encephalomyelitis at age 90, still relevant and elucidating how viruses trigger disease. J Exp Med 2023; 220:213807. [PMID: 36652203 PMCID: PMC9880878 DOI: 10.1084/jem.20221322] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/28/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
20 yr ago, a tribute appeared in this journal on the 70th anniversary of an animal model of disseminated encephalomyelitis, abbreviated EAE for experimental autoimmune encephalomyelitis. "Observations on Attempts to Produce Disseminated Encephalomyelitis in Monkeys" appeared in the Journal of Experimental Medicine on February 21, 1933. Rivers and colleagues were trying to understand what caused neurological reactions to viral infections like smallpox, vaccinia, and measles, and what triggered rare instances of encephalomyelitis to smallpox vaccines. The animal model known as EAE continues to display its remarkable utility. Recent research, since the 70th-anniversary tribute, helps explain how Epstein-Barr virus triggers multiple sclerosis via molecular mimicry to a protein known as GlialCAM. Proteins with multiple domains similar to GlialCAM, tenascin, neuregulin, contactin, and protease kinase C inhibitors are present in the poxvirus family. These observations take us a full circle back to Rivers' first paper on EAE, 90 yr ago.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences and Pediatrics, Stanford University, Stanford, CA, USA,Correspondence to Lawrence Steinman:
| | | | | |
Collapse
|
6
|
Höftberger R, Lassmann H, Berger T, Reindl M. Pathogenic autoantibodies in multiple sclerosis - from a simple idea to a complex concept. Nat Rev Neurol 2022; 18:681-688. [PMID: 35970870 DOI: 10.1038/s41582-022-00700-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
The role of autoantibodies in multiple sclerosis (MS) has been enigmatic since the first description, many decades ago, of intrathecal immunoglobulin production in people with this condition. Some studies have indicated that MS pathology is heterogeneous, with an antibody-associated subtype - characterized by B cells (in varying quantities), antibodies and complement - existing alongside other subtypes with different pathologies. However, subsequent evidence suggested that some cases originally diagnosed as MS with autoantibody-mediated demyelination were more likely to be neuromyelitis optica spectrum disorder or myelin oligodendrocyte glycoprotein antibody-associated disease. These findings raise the important question of whether an autoantibody-mediated MS subtype exists and whether pathogenic MS-associated autoantibodies remain to be identified. Potential roles of autoantibodies in MS could range from specific antibodies defining the disease to a non-disease-specific amplification of cellular immune responses and other pathophysiological processes. In this Perspective, we review studies that have attempted to identify MS-associated autoantibodies and provide our opinions on their possible roles in the pathophysiology of MS.
Collapse
Affiliation(s)
- Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
7
|
Affiliation(s)
- Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA 02215
- Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
8
|
Martín-Márquez BT, Satoh M, Hernández-Pando R, Martínez-García EA, Petri MH, Sandoval-García F, Pizano-Martinez O, García-Iglesias T, Corona-Meraz FI, Vázquez-Del Mercado M. The DNA co-vaccination using Sm antigen and IL-10 as prophylactic experimental therapy ameliorates nephritis in a model of lupus induced by pristane. PLoS One 2021; 16:e0259114. [PMID: 34705865 PMCID: PMC8550422 DOI: 10.1371/journal.pone.0259114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies such as anti-Sm. Studies in patients with SLE and murine models of lupus reveal that the most critical anti-Sm autoantibodies are predominantly direct against D1(83–119), D2, and B´/B epitopes. Objectives The present study aimed to analyze the induction of antigen-specific tolerance after prophylactic immunization with a DNA vaccine encoding the epitopes: D183-119, D2, B´/B, and B´/BCOOH in co-vaccination with IFN-γ or IL-10 in a murine model of lupus induced by pristane. Material and methods To obtain endotoxin-free DNA vaccines, direct cloning techniques using pcDNA were performed: D183-119, D2, B´/B, B´/BCOOH, IFN-γ, or IL-10. Lupus was induced by 0.5 mL of pristane via intraperitoneal in BALB/c female mice. Immunoprecipitation with K562 cells was metabolically labeled with 35S and ELISA to detect serum antibodies or mice IgG1, IgG2a isotypes. ELISA determined IL-10 and IFN-γ from splenocytes supernatants. Proteinuria was assessed monthly, and lupus nephritis was evaluated by immunofluorescence, and electron microscopy. Results The prophylactic co-vaccination with D2/IL-10 reduced the expression of kidney damage observed by electron microscopy, direct immunofluorescence, and H & E, along with reduced level of anti-nRNP/Sm antibodies (P = 0.048). Conclusion The prophylactic co-vaccination of IL-10 with D2 in pristane-induced lupus ameliorates the renal damage maybe by acting as prophylactic DNA tolerizing therapy.
Collapse
Affiliation(s)
- Beatriz Teresita Martín-Márquez
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-703, "Inmunología y Reumatología", Guadalajara, Mexico
| | - Minoru Satoh
- Department of Clinical Nursing, School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Rogelio Hernández-Pando
- Departamento de Patología, Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Erika Aurora Martínez-García
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-703, "Inmunología y Reumatología", Guadalajara, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Marcelo Heron Petri
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Department of Cardiothoracic and Vascular Surgery, Örebro University Hospital, Örebro, Sweden
| | - Flavio Sandoval-García
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-701, "Envejecimiento, Inmunometabolismo y estrés oxidativo", Ciudad de La Habana, Cuba
| | - Oscar Pizano-Martinez
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-703, "Inmunología y Reumatología", Guadalajara, Mexico
- Departamento de Morfología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Trinidad García-Iglesias
- Departamento de Fisiología, Laboratorio de Inmunología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Fernanda Isadora Corona-Meraz
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-701, "Envejecimiento, Inmunometabolismo y estrés oxidativo", Ciudad de La Habana, Cuba
- División de Ciencias de la Salud, Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá, Jalisco, México
| | - Monica Vázquez-Del Mercado
- Departamento de Biología Molecular, Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético (IIRSME), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
- Departamento de Biología Molecular, UDG-CA-703, "Inmunología y Reumatología", Guadalajara, Mexico
- División de Medicina Interna, Hospital Civil "Dr. Juan I. Menchaca", Servicio de Reumatología PNPC 004086 CONACyT, Guadalajara, Jalisco, México
| |
Collapse
|
9
|
Autoantibodies against central nervous system antigens in a subset of B cell-dominant multiple sclerosis patients. Proc Natl Acad Sci U S A 2020; 117:21512-21518. [PMID: 32817492 DOI: 10.1073/pnas.2011249117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), with characteristic inflammatory lesions and demyelination. The clinical benefit of cell-depleting therapies targeting CD20 has emphasized the role of B cells and autoantibodies in MS pathogenesis. We previously introduced an enzyme-linked immunospot spot (ELISpot)-based assay to measure CNS antigen-specific B cells in the blood of MS patients and demonstrated its usefulness as a predictive biomarker for disease activity in measuring the successful outcome of disease-modifying therapies (DMTs). Here we used a planar protein array to investigate CNS-reactive antibodies in the serum of MS patients as well as in B cell culture supernatants after polyclonal stimulation. Anti-CNS antibody reactivity was evident in the sera of the MS cohort, and the antibodies bound a heterogeneous set of molecules, including myelin, axonal cytoskeleton, and ion channel antigens, in individual patients. Immunoglobulin reactivity in supernatants of stimulated B cells was directed against a broad range of CNS antigens. A group of MS patients with a highly active B cell component was identified by the ELISpot assay. Those antibody reactivities remained stable over time. These assays with protein arrays identify MS patients with a highly active B cell population with antibodies directed against a swathe of CNS proteins.
Collapse
|
10
|
The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 2019; 19:184-197. [PMID: 30718831 DOI: 10.1038/s41577-019-0125-8] [Citation(s) in RCA: 778] [Impact Index Per Article: 129.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.
Collapse
|
11
|
Taghavian O, Jain A, Joyner CJ, Ketchum S, Nakajima R, Jasinskas A, Liang L, Fong R, King C, Greenhouse B, Murphy M, Bailey J, Galinski MR, Barnwell JW, Plowe CV, Davies DH, Felgner PL. Antibody Profiling by Proteome Microarray with Multiplex Isotype Detection Reveals Overlap between Human and Aotus nancymaae Controlled Malaria Infections. Proteomics 2019; 18. [PMID: 29266845 DOI: 10.1002/pmic.201700277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/21/2017] [Indexed: 12/13/2022]
Abstract
The development of vaccines against malaria and serodiagnostic tests for detecting recent exposure requires tools for antigen discovery and suitable animal models. The protein microarray is a high-throughput, sample sparing technique, with applications in infectious disease research, clinical diagnostics, epidemiology, and vaccine development. We recently demonstrated Qdot-based indirect immunofluorescence together with portable optical imager ArrayCAM using single isotype detection could replicate data using the conventional laser confocal scanner system. We developed a multiplexing protocol for simultaneous detection of IgG, IgA, and IgM and compared samples from a controlled human malaria infection model with those from controlled malaria infections of Aotus nancymaae, a widely used non-human primate model of human malaria. IgG profiles showed the highest concordance in number of reactive antigens; thus, of the 139 antigens recognized by human IgG antibody, 111 were also recognized by Aotus monkeys. Interestingly, IgA profiles were largely non-overlapping. Finally, on the path toward wider deployment of the portable platform, we show excellent correlations between array data obtained in five independent laboratories around the United States using the multiplexing protocol (R2 : 0.60-0.92). This study supports the use of this platform for wider deployment, particularly in endemic areas where such a tool will have the greatest impact on global human health.
Collapse
Affiliation(s)
- Omid Taghavian
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Aarti Jain
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Chester J Joyner
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Rie Nakajima
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Algis Jasinskas
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Li Liang
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Rich Fong
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Maxwell Murphy
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jason Bailey
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mary R Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - John W Barnwell
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christopher V Plowe
- Howard Hughes Medical Institute/Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - D Huw Davies
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Philip L Felgner
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Engineered DNA plasmid reduces immunity to dystrophin while improving muscle force in a model of gene therapy of Duchenne dystrophy. Proc Natl Acad Sci U S A 2018; 115:E9182-E9191. [PMID: 30181272 DOI: 10.1073/pnas.1808648115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In gene therapy for Duchenne muscular dystrophy there are two potential immunological obstacles. An individual with Duchenne muscular dystrophy has a genetic mutation in dystrophin, and therefore the wild-type protein is "foreign," and thus potentially immunogenic. The adeno-associated virus serotype-6 (AAV6) vector for delivery of dystrophin is a viral-derived vector with its own inherent immunogenicity. We have developed a technology where an engineered plasmid DNA is delivered to reduce autoimmunity. We have taken this approach into humans, tolerizing to myelin proteins in multiple sclerosis and to proinsulin in type 1 diabetes. Here, we extend this technology to a model of gene therapy to reduce the immunogenicity of the AAV vector and of the wild-type protein product that is missing in the genetic disease. Following gene therapy with systemic administration of recombinant AAV6-microdystrophin to mdx/mTRG2 mice, we demonstrated the development of antibodies targeting dystrophin and AAV6 capsid in control mice. Treatment with the engineered DNA construct encoding microdystrophin markedly reduced antibody responses to dystrophin and to AAV6. Muscle force in the treated mice was also improved compared with control mice. These data highlight the potential benefits of administration of an engineered DNA plasmid encoding the delivered protein to overcome critical barriers in gene therapy to achieve optimal functional gene expression.
Collapse
|
13
|
Qendro V, Bugos GA, Lundgren DH, Glynn J, Han MH, Han DK. Integrative proteomics, genomics, and translational immunology approaches reveal mutated forms of Proteolipid Protein 1 (PLP1) and mutant-specific immune response in multiple sclerosis. Proteomics 2017; 17. [PMID: 28191734 DOI: 10.1002/pmic.201600322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/28/2017] [Accepted: 02/08/2017] [Indexed: 12/14/2022]
Abstract
In order to gain mechanistic insights into multiple sclerosis (MS) pathogenesis, we utilized a multi-dimensional approach to test the hypothesis that mutations in myelin proteins lead to immune activation and central nervous system autoimmunity in MS. Mass spectrometry-based proteomic analysis of human MS brain lesions revealed seven unique mutations of PLP1; a key myelin protein that is known to be destroyed in MS. Surprisingly, in-depth genomic analysis of two MS patients at the genomic DNA and mRNA confirmed mutated PLP1 in RNA, but not in the genomic DNA. Quantification of wild type and mutant PLP RNA levels by qPCR further validated the presence of mutant PLP RNA in the MS patients. To seek evidence linking mutations in abundant myelin proteins and immune-mediated destruction of myelin, specific immune response against mutant PLP1 in MS patients was examined. Thus, we have designed paired, wild type and mutant peptide microarrays, and examined antibody response to multiple mutated PLP1 in sera from MS patients. Consistent with the idea of different patients exhibiting unique mutation profiles, we found that 13 out of 20 MS patients showed antibody responses against specific but not against all the mutant-PLP1 peptides. Interestingly, we found mutant PLP-directed antibody response against specific mutant peptides in the sera of pre-MS controls. The results from integrative proteomic, genomic, and immune analyses reveal a possible mechanism of mutation-driven pathogenesis in human MS. The study also highlights the need for integrative genomic and proteomic analyses for uncovering pathogenic mechanisms of human diseases.
Collapse
Affiliation(s)
- Veneta Qendro
- Department of Cell Biology, Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Grace A Bugos
- Department of Cell Biology, Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Debbie H Lundgren
- Department of Cell Biology, Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - John Glynn
- Molecular Core Facility, University of Connecticut School of Medicine, Farmington, CT, USA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Han
- Department of Cell Biology, Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
14
|
Nazeen S, Palmer NP, Berger B, Kohane IS. Integrative analysis of genetic data sets reveals a shared innate immune component in autism spectrum disorder and its co-morbidities. Genome Biol 2016; 17:228. [PMID: 27842596 PMCID: PMC5108086 DOI: 10.1186/s13059-016-1084-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 10/12/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a common neurodevelopmental disorder that tends to co-occur with other diseases, including asthma, inflammatory bowel disease, infections, cerebral palsy, dilated cardiomyopathy, muscular dystrophy, and schizophrenia. However, the molecular basis of this co-occurrence, and whether it is due to a shared component that influences both pathophysiology and environmental triggering of illness, has not been elucidated. To address this, we deploy a three-tiered transcriptomic meta-analysis that functions at the gene, pathway, and disease levels across ASD and its co-morbidities. RESULTS Our analysis reveals a novel shared innate immune component between ASD and all but three of its co-morbidities that were examined. In particular, we find that the Toll-like receptor signaling and the chemokine signaling pathways, which are key pathways in the innate immune response, have the highest shared statistical significance. Moreover, the disease genes that overlap these two innate immunity pathways can be used to classify the cases of ASD and its co-morbidities vs. controls with at least 70 % accuracy. CONCLUSIONS This finding suggests that a neuropsychiatric condition and the majority of its non-brain-related co-morbidities share a dysregulated signal that serves as not only a common genetic basis for the diseases but also as a link to environmental triggers. It also raises the possibility that treatment and/or prophylaxis used for disorders of innate immunity may be successfully used for ASD patients with immune-related phenotypes.
Collapse
Affiliation(s)
- Sumaiya Nazeen
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139 MA USA
| | - Nathan P. Palmer
- Department of Biomedical Informatics, Harvard Medical School, 25 Shattuck Street, Boston, 02115 MA USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139 MA USA
- Department of Mathematics, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139 MA USA
| | - Isaac S. Kohane
- Department of Biomedical Informatics, Harvard Medical School, 25 Shattuck Street, Boston, 02115 MA USA
| |
Collapse
|
15
|
Steinman L. A Journey in Science: The Privilege of Exploring the Brain and the Immune System. Mol Med 2016; 22:molmed.2015.00263. [PMID: 27652378 PMCID: PMC5004718 DOI: 10.2119/molmed.2015.00263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/06/2022] Open
Abstract
Real innovations in medicine and science are historic and singular; the stories behind each occurrence are precious. At Molecular Medicine we have established the Anthony Cerami Award in Translational Medicine to document and preserve these histories. The monographs recount the seminal events as told in the voice of the original investigators who provided the crucial early insight. These essays capture the essence of discovery, chronicling the birth of ideas that created new fields of research; and launched trajectories that persisted and ultimately influenced how disease is prevented, diagnosed, and treated. In this volume, the Cerami Award Monograph is by Lawrence Steinman, MD, of Stanford University in California. A visionary in the field of neurology, this is the story of Dr. Steinman's scientific journey.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| |
Collapse
|
16
|
Anoctamin 2 identified as an autoimmune target in multiple sclerosis. Proc Natl Acad Sci U S A 2016; 113:2188-93. [PMID: 26862169 DOI: 10.1073/pnas.1518553113] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory disease of the central nervous system and also is regarded as an autoimmune condition. However, the antigenic targets of the autoimmune response in MS have not yet been deciphered. In an effort to mine the autoantibody repertoire within MS, we profiled 2,169 plasma samples from MS cases and population-based controls using bead arrays built with 384 human protein fragments selected from an initial screening with 11,520 antigens. Our data revealed prominently increased autoantibody reactivity against the chloride-channel protein anoctamin 2 (ANO2) in MS cases compared with controls. This finding was corroborated in independent assays with alternative protein constructs and by epitope mapping with peptides covering the identified region of ANO2. Additionally, we found a strong interaction between the presence of ANO2 autoantibodies and the HLA complex MS-associated DRB1*15 allele, reinforcing a potential role for ANO2 autoreactivity in MS etiopathogenesis. Furthermore, immunofluorescence analysis in human MS brain tissue showed ANO2 expression as small cellular aggregates near and inside MS lesions. Thus this study represents one of the largest efforts to characterize the autoantibody repertoire within MS. The findings presented here demonstrate that an ANO2 autoimmune subphenotype may exist in MS and lay the groundwork for further studies focusing on the pathogenic role of ANO2 autoantibodies in MS.
Collapse
|
17
|
A Liposome-Based Approach to the Integrated Multi-Component Antigen Microarrays. MICROARRAYS 2015; 4:618-29. [PMID: 27600243 PMCID: PMC4996415 DOI: 10.3390/microarrays4040618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022]
Abstract
This report describes an experimental procedure for constructing integrated lipid, carbohydrate, and protein microarrays. In essence, it prints liposomes on nitrocellulose-coated micro-glass slides, a biochip substrate for spotting protein and carbohydrate microarrays, and the substances that can form liposomes (homo-liposomes) or can be incorporated into liposomes (hetero-liposomes) are suitable for microarray construction using existing microarray spotting devices. Importantly, this technology allows simultaneous detection of serum antibody activities among the three major classes of antigens, i.e., lipids, carbohydrates, and proteins. The potential of this technology is illustrated by its use in revealing a broad-spectrum of pre-existing anti-lipid antibodies in blood circulation and monitoring the epitope spreading of autoantibody reactivities among protein, carbohydrate, and lipid antigens in experimental autoimmune encephalomyelitis (EAE).
Collapse
|
18
|
Doran TM, Gao Y, Simanski S, McEnaney P, Kodadek T. High affinity binding of conformationally constrained synthetic oligomers to an antigen-specific antibody: Discovery of a diagnostically useful synthetic ligand for murine Type 1 diabetes autoantibodies. Bioorg Med Chem Lett 2015; 25:4910-4917. [PMID: 26067174 PMCID: PMC4607566 DOI: 10.1016/j.bmcl.2015.05.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 05/18/2015] [Indexed: 10/23/2022]
Abstract
'Antigen surrogates' are synthetic, non-natural molecules that recognize the antigen-binding sites of antibodies. These molecules are of interest as replacements for native antigens as antibody 'capture agents' in ELISA-like assays of potential diagnostic utility, for example when the antibody is indicative of a disease state. Antigen surrogates for disease-related antibodies can be mined from one-bead one-compound (OBOC) libraries by first denuding the library of ligands for antibodies present in the serum of control patients or animals, followed by screening the remainder of the library against serum from individuals with a particular disease of interest. Most of the work in this area has been done with peptoids (oligomers of N-alkylated glycine), which provide antibody ligands with only modest affinity and selectivity. Here, we explore the hypothesis that this is due to the 'floppiness' of the peptoid backbone by creating libraries of peptoid-like molecules that have conformation-restricting structural elements inserted into their backbones. Indeed, we show here that these libraries can provide high affinity and selectivity antigen surrogates and that this much-improved binding is completely dependent on conformational restriction of the oligomer chain.
Collapse
Affiliation(s)
- Todd M Doran
- The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Yu Gao
- The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Scott Simanski
- The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Patrick McEnaney
- The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Thomas Kodadek
- The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, United States.
| |
Collapse
|
19
|
Ligocki AJ, Rivas JR, Rounds WH, Guzman AA, Li M, Spadaro M, Lahey L, Chen D, Henson PM, Graves D, Greenberg BM, Frohman EM, Ward ES, Robinson W, Meinl E, White CL, Stowe AM, Monson NL. A Distinct Class of Antibodies May Be an Indicator of Gray Matter Autoimmunity in Early and Established Relapsing Remitting Multiple Sclerosis Patients. ASN Neuro 2015; 7:7/5/1759091415609613. [PMID: 26489686 PMCID: PMC4710131 DOI: 10.1177/1759091415609613] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
*These authors contributed equally to the work in this manuscript.We have previously identified a distinct class of antibodies expressed by B cells in the cerebrospinal fluid (CSF) of early and established relapsing remitting multiple sclerosis (RRMS) patients that is not observed in healthy donors. These antibodies contain a unique pattern of mutations in six codons along VH4 antibody genes that we termed the antibody gene signature (AGS). In fact, patients who have such B cells in their CSF are identified as either having RRMS or developing RRMS in the future. As mutations in antibody genes increase antibody affinity for particular antigens, the goal for this study was to investigate whether AGS(+) antibodies bind to brain tissue antigens. Single B cells were isolated from the CSF of 10 patients with early or established RRMS. We chose 32 of these B cells that expressed antibodies enriched for the AGS for further study. We generated monoclonal full-length recombinant human antibodies (rhAbs) and used both immunological assays and immunohistochemistry to investigate the capacity of these AGS(+) rhAbs to bind brain tissue antigens. AGS(+) rhAbs did not recognize myelin tracts in the corpus callosum. Instead, AGS(+) rhAbs recognized neuronal nuclei and/or astrocytes, which are prevalent in the cortical gray matter. This pattern was unique to the AGS(+) antibodies from early and established RRMS patients, as AGS(+) antibodies from an early neuromyelitis optica patient did not display the same reactivity. Prevalence of CSF-derived B cells expressing AGS(+) antibodies that bind to these cell types may be an indicator of gray matter-directed autoimmunity in early and established RRMS patients.
Collapse
Affiliation(s)
- Ann J Ligocki
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jacqueline R Rivas
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William H Rounds
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alyssa A Guzman
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Min Li
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melania Spadaro
- Institute of Clinical Neuroimmunology, Ludwig-Maximilian-University, Munich, Germany
| | - Lauren Lahey
- Department of Immunology and Rheumatology, Stanford University, CA, USA
| | - Ding Chen
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Paul M Henson
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donna Graves
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin M Greenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elliot M Frohman
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William Robinson
- Department of Immunology and Rheumatology, Stanford University, CA, USA
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Ludwig-Maximilian-University, Munich, Germany
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Farjam M, Zhang GX, Ciric B, Rostami A. Emerging immunopharmacological targets in multiple sclerosis. J Neurol Sci 2015; 358:22-30. [PMID: 26440421 DOI: 10.1016/j.jns.2015.09.346] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 10/23/2022]
Abstract
Inflammatory demyelination of the central nervous system (CNS) is the hallmark of multiple sclerosis (MS), a chronic debilitating disease that affects more than 2.5 million individuals worldwide. It has been widely accepted, although not proven, that the major pathogenic mechanism of MS involves myelin-reactive T cell activation in the periphery and migration into the CNS, which subsequently triggers an inflammatory cascade that leads to demyelination and axonal damage. Virtually all MS medications now in use target the immune system and prevent tissue damage by modulating neuroinflammatory processes. Although current therapies such as commonly prescribed disease-modifying medications decrease the relapse rate in relapsing-remitting MS (RRMS), the prevention of long-term accumulation of deficits remains a challenge. Medications used for progressive forms of MS also have limited efficacy. The need for therapies that are effective against disease progression continues to drive the search for novel pharmacological targets. In recent years, due to a better understanding of MS immunopathogenesis, new approaches have been introduced that more specifically target autoreactive immune cells and their products, thus increasing specificity and efficacy, while reducing potential side effects such as global immunosuppression. In this review we describe several immunopharmacological targets that are currently being explored for MS therapy.
Collapse
Affiliation(s)
- Mojtaba Farjam
- Non-communicable Diseases Research Center, Department of Medical Pharmacology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
21
|
Furman D, Davis MM. New approaches to understanding the immune response to vaccination and infection. Vaccine 2015; 33:5271-81. [PMID: 26232539 DOI: 10.1016/j.vaccine.2015.06.117] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/26/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
The immune system is a network of specialized cell types and tissues that communicates via cytokines and direct contact, to orchestrate specific types of defensive responses. Until recently, we could only study immune responses in a piecemeal, highly focused fashion, on major components like antibodies to the pathogen. But recent advances in technology and in our understanding of the many components of the system, innate and adaptive, have made possible a broader approach, where both the multiple responding cells and cytokines in the blood are measured. This systems immunology approach to a vaccine response or an infection gives us a more holistic picture of the different parts of the immune system that are mobilized and should allow us a much better understanding of the pathways and mechanisms of such responses, as well as to predict vaccine efficacy in different populations well in advance of efficacy studies. Here we summarize the different technologies and methods and discuss how they can inform us about the differences between diseases and vaccines, and how they can greatly accelerate vaccine development.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States; Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
22
|
Berger T, Reindl M. Antibody biomarkers in CNS demyelinating diseases - a long and winding road. Eur J Neurol 2015; 22:1162-8. [PMID: 26010364 DOI: 10.1111/ene.12759] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/27/2015] [Indexed: 01/13/2023]
Abstract
Over several decades, studies sought potential markers to diagnose and to predict the clinical course of central nervous system (CNS) demyelinating disorders, especially in multiple sclerosis, acute disseminated encephalomyelitis and neuromyelitis optica spectrum disorders. Reliable biomarkers would ensure correct diagnoses, determine future disease evolvements, stratify patients for appropriate treatments and monitor disease activity and treatment effects - in summary, meet the longing for personalized medicine in these diseases. Out of a plethora of potential biomarker candidates antibodies have turned (again) into the scientific focus, due to pivotal immunological and neuropathological findings in the past 20 years. A major breakthrough and stimulus for further research was the identification of anti-aquaporin-4 antibodies in neuromyelitis optica. Various other myelin and non-myelin antigens were investigated in detail for diagnostic and prognostic purposes, such as antibodies to myelin oligodendrocyte glycoprotein or to the potassium channel KIR4.1. Further, the use of biopharmaceutical treatments in multiple sclerosis led to intense research activities to identify anti-treatment neutralizing antibodies and their clinical consequences. This review briefly summarizes the current knowledge on antibodies in the diagnosis, prognosis, disease and treatment monitoring of CNS demyelinating disorders.
Collapse
Affiliation(s)
- T Berger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - M Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
Recks MS, Grether NB, van der Broeck F, Ganscher A, Wagner N, Henke E, Ergün S, Schroeter M, Kuerten S. Four different synthetic peptides of proteolipid protein induce a distinct antibody response in MP4-induced experimental autoimmune encephalomyelitis. Clin Immunol 2015; 159:93-106. [PMID: 25959684 DOI: 10.1016/j.clim.2015.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 04/30/2015] [Indexed: 01/26/2023]
Abstract
Here we studied the autoantibody specificity elicited by proteolipid protein (PLP) in MP4-induced experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis (MS). In C57BL/6 (B6) mice, antibodies were induced by immunization with one of the two extracellular and by the intracellular PLP domain. Antibodies against extracellular PLP were myelin-reactive in oligodendrocyte cultures and induced mild spinal cord demyelination upon transfer into B cell-deficient J(H)T mice. Remarkably, also antibodies against intracellular PLP showed binding to intact oligodendrocytes and were capable of inducing myelin pathology upon transfer into J(H)T mice. In MP4-immunized mice peptide-specific T(H)1/T(H)17 responses were mainly directed against the extracellular PLP domains, but also involved the intracellular epitopes. These data suggest that both extracellular and intracellular epitopes of PLP contribute to the pathogenesis of MP4-induced EAE already in the setting of intact myelin. It remains to be elucidated if this concept also applies to MS itself.
Collapse
Affiliation(s)
- Mascha S Recks
- Department of Anatomy II (Neuroanatomy), University of Cologne, Kerpener Straβe 62, 50924 Cologne, Germany
| | - Nicolai B Grether
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | | | - Alla Ganscher
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Nicole Wagner
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Erik Henke
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Süleyman Ergün
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany
| | - Michael Schroeter
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50924 Cologne, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstr. 6, 97070 Wuerzburg, Germany.
| |
Collapse
|
24
|
Huang QQ, Perlman H, Birkett R, Doyle R, Fang D, Haines GK, Robinson W, Datta S, Huang Z, Li QZ, Phee H, Pope RM. CD11c-mediated deletion of Flip promotes autoreactivity and inflammatory arthritis. Nat Commun 2015; 6:7086. [PMID: 25963626 PMCID: PMC4429912 DOI: 10.1038/ncomms8086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are critical for immune homeostasis. To target DCs, we generated a mouse line with Flip deficiency in cells that express cre under the CD11c promoter (CD11c-Flip-KO). CD11c-Flip-KO mice spontaneously develop erosive, inflammatory arthritis, resembling rheumatoid arthritis, which is dramatically reduced when these mice are crossed with Rag−/− mice. The CD8α+ DC subset is significantly reduced, along with alterations in NK cells and macrophages. Autoreactive CD4+ T cells and autoantibodies specific for joint tissue are present, and arthritis severity correlates with the number of autoreactive CD4+ T cells and plasmablasts in the joint-draining lymph nodes. Reduced T regulatory cells (Tregs) inversely correlate with arthritis severity, and the transfer of Tregs ameliorates arthritis. This KO line identifies a model that will permit in depth interrogation of the pathogenesis of rheumatoid arthritis, including the role of CD8α+ DCs and other cells of the immune system. Dendritic cells are critical for initiation of immune responses and for induction of tolerance. Here the authors show that deletion of survival factor c-flip in CD11c-expressing cells subset perturbs CD8a+ dendritic cell, NK and macrophage pools, and leads to development of autoimmune arthritis.
Collapse
Affiliation(s)
- Qi-Quan Huang
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Harris Perlman
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Robert Birkett
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Renee Doyle
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - G Kenneth Haines
- Department of Pathology, Mount Sinai Hospital School of Medicine, New York city, New York 10029, USA
| | - William Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, VA Health Care System, Palo Alto, California 94304, USA
| | - Syamal Datta
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Zan Huang
- Department of Biochemistry, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Hyewon Phee
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Richard M Pope
- Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
25
|
Abstract
Ideal therapy for inflammatory disease in the nervous system would preserve normal immune function, while suppressing only the pathologic immune responses that damage tissue and allowing for repair. In principle, antigen-specific therapy would eradicate unwanted adaptive immune responses-antibody and T-cell mediated--while preserving the integrity of other adaptive responses to infectious agents and retaining the ability to fight malignancy. However, at this time, for multiple sclerosis (MS) we do not have compelling evidence that would support any particular dominant immune response to any specific antigen or even a limited group of antigens. In fact, there are adaptive immune responses to a wide swathe of proteins and lipids found on neurons and myelin in MS. Unless controlling a few of the known immune responses is sufficient, antigen-specific therapy in MS may not have enough of an impact to modulate clinical outcome. However, in other neuroinflammatory conditions, such as neuromyelitis optica, the adaptive immune response is highly focused. Trials of antigen-specific therapy for neuroinflammatory disease might first be tested in diseases with a more limited adaptive immune response like neuromyelitis optica. The likelihood of a significant success for this therapeutic strategy might then ensue.
Collapse
|
26
|
Rosenberg JM, Utz PJ. Protein microarrays: a new tool for the study of autoantibodies in immunodeficiency. Front Immunol 2015; 6:138. [PMID: 25904912 PMCID: PMC4387933 DOI: 10.3389/fimmu.2015.00138] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/12/2015] [Indexed: 12/19/2022] Open
Abstract
Autoimmunity is highly coincident with immunodeficiency. In a small but growing number of primary immunodeficiencies, autoantibodies are diagnostic of a given disease and implicated in disease pathogenesis. In order to improve our understanding of the role of autoantibodies in immunodeficiencies and to discover novel autoantibodies, new proteomic tools are needed. Protein microarrays have the ability to screen for reactivity to hundreds to many thousands of unique autoantigens simultaneously on a single chip using minimal serum input. Here, we review different types of protein microarrays and how they can be useful in framing the study of primary and secondary immunodeficiencies.
Collapse
Affiliation(s)
- Jacob M Rosenberg
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine , Stanford, CA , USA
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine , Stanford, CA , USA ; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
27
|
Development of therapies for autoimmune disease at Stanford: a tale of multiple shots and one goal. Immunol Res 2015; 58:307-14. [PMID: 24771483 DOI: 10.1007/s12026-014-8509-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The title of this contribution on Immunology at Stanford is purposely ambiguous. One goal is the development of safe and effective therapy for autoimmune diseases. Another definition of goal is to score, and this would ultimately mean the development of an approved drug. Indeed, the efforts in my four decades at Stanford, have included the discovery and subsequent development of a monoclonal antibody to block homing to the inflamed brain, leading to natalizumab, an approved therapeutic for two autoimmune diseases: relapsing-remitting MS and for inflammatory bowel disease. Multiple attempts to develop new therapies for autoimmune disease are described here: The trimolecular complex and the immune synapse serve as one major set of targets, with attempts to inhibit particular major histocompatibility molecules, the variable regions of the T cell receptor, and CD4. Other approaches focusing on antigen-specific tolerance include ongoing attempts with tolerizing DNA vaccines in type 1 diabetes. Finally, the repurposing of popular drugs approved for other indications, including statins and inhibitors of angiotensin converting enzyme is under development and showing promise in the clinic, particularly for secondary progressive multiple sclerosis. The milieu within Stanford Immunology has helped to nurture these efforts to translate discoveries in immunology and to take them from bench to bedside.
Collapse
|
28
|
Abstract
ABSTRACT:The adaptive immune response in multiple sclerosis is complex. We have devised large scale arrays to measure the antibody response to myelin proteins and lipids. Despite the widespread immune responses to myelin, we have devised an inverse vaccine aimed at turning off key drivers of this diverse response. Clinical trials in patients with multiple sclerosis show that it is possible to constrain antibody responses to myelin on a large scale with this approach.
Collapse
|
29
|
Quintana FJ, Patel B, Yeste A, Nyirenda M, Kenison J, Rahbari R, Fetco D, Hussain M, O'Mahony J, Magalhaes S, McGowan M, Johnson T, Rajasekharan S, Narayanan S, Arnold DL, Weiner HL, Banwell B, Bar-Or A. Epitope spreading as an early pathogenic event in pediatric multiple sclerosis. Neurology 2014; 83:2219-26. [PMID: 25381299 DOI: 10.1212/wnl.0000000000001066] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES For most adults with initial clinical presentation of multiple sclerosis (MS), biological disease was likely initiated many years prior. Pediatric-onset MS provides an opportunity to study early disease processes. METHODS Using antigen microarrays, including CNS-related proteins, lipids, and other autoantigens, we studied early immunologic events involved in clinical onset of pediatric MS. Serum samples were collected at the time of incident acquired CNS demyelinating syndromes (ADS) in children who, in subsequent prospective follow-up, were ascertained to have either pediatric MS (ADS-MS) or a monophasic illness (ADS-mono). Samples were obtained both at the time of ADS presentation and 3 months into follow-up. We used an initial training set of samples to implicate antibody signatures associated with each group, and then a test set. An additional set of follow-up samples (stability set) was used as a form of internal validation. RESULTS Children with ADS-MS tended to have distinguishable serum antibody patterns both at the time of ADS presentation and 3 months into follow-up. At the time of ADS, serum samples from patients with ADS-MS or ADS-mono reacted against similar numbers of CNS antigens, although CNS antigens implicated in adult MS were more often targeted in children with ADS-MS. The follow-up ADS-MS samples reacted against a broader panel of CNS antigens, while corresponding ADS-mono samples exhibited a contraction of the initial antibody response. CONCLUSIONS Our findings in this prospective cohort of pediatric-onset CNS demyelinating diseases point to an active process of epitope spreading during early stages of MS, not seen in monophasic CNS inflammatory conditions.
Collapse
Affiliation(s)
- Francisco J Quintana
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Bonny Patel
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Ada Yeste
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Mukanthu Nyirenda
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Jessica Kenison
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Roya Rahbari
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Dumitru Fetco
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Mohammad Hussain
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Julia O'Mahony
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sandra Magalhaes
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Melissa McGowan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Trina Johnson
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sathy Rajasekharan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sridar Narayanan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Douglas L Arnold
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Howard L Weiner
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Brenda Banwell
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Amit Bar-Or
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA.
| | | |
Collapse
|
30
|
Steinman L, Shoenfeld Y. From defining antigens to new therapies in multiple sclerosis: honoring the contributions of Ruth Arnon and Michael Sela. J Autoimmun 2014; 54:1-7. [PMID: 25308417 DOI: 10.1016/j.jaut.2014.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 08/12/2014] [Indexed: 12/21/2022]
Abstract
Ruth Arnon and Michael Sela profoundly influenced the development of a model system to test new therapies in multiple sclerosis (MS). Their application of the animal model, known as experimental autoimmune encephalomyelitis (EAE), for the discovery of Copaxone, opened a new path for testing of drug candidates in MS. By measuring clinical, pathologic, and immunologic outcomes, the biological implications of new drugs could be elucidated. Using EAE they established the efficacy of Copaxone as a therapy for preventing and reducing paralysis and inflammation in the central nervous system without massive immune suppression. This had a huge impact on the field of drug discovery for MS. Much like the use of parabiosis to discover soluble factors associated with obesity, or the replica plating system to probe antibiotic resistance in bacteria, the pioneering research on Copaxone using the EAE model, paved the way for the discovery of other therapeutics in MS, including Natalizumab and Fingolimod. Future applications of this approach may well elucidate novel therapies for the neurodegenerative phase of multiple sclerosis associated with disease progression.
Collapse
Affiliation(s)
- Lawrence Steinman
- Beckman Center for Molecular Medicine, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
31
|
Notturno F, Di Febo T, Yuki N, Fernandez Rodriguez BM, Corti D, Nobile-Orazio E, Carpo M, De Lauretis A, Uncini A. Autoantibodies to neurofascin-186 and gliomedin in multifocal motor neuropathy. J Neuroimmunol 2014; 276:207-12. [PMID: 25283719 DOI: 10.1016/j.jneuroim.2014.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 11/17/2022]
Abstract
We tested autoantibodies to neurofascin-186 (NF186) and gliomedin in sera from patients with multifocal motor neuropathy (MMN, n=53) and chronic inflammatory demyelinating polyneuropathy (CIDP, n=95) by ELISA. IgG antibodies to NF186 or gliomedin were found in 62% of MMN and 1% of CIDP sera, and IgM antibodies to the same antigens in 12% of MMN and 1% of CIDP sera. These autoantibodies activated complement. Ten percent of the MMN sera without IgM anti-GM1 reactivity had anti-NF186 antibodies. Because NF186 and gliomedin play a crucial role for salutatory conduction, the autoantibodies may contribute to produce motor nerve conduction block and muscle weakness in MMN.
Collapse
Affiliation(s)
- Francesca Notturno
- Department of Neuroscience and Imaging, University "G. d'Annunzio", Chieti-Pescara, Italy.
| | - Tiziana Di Febo
- Experimental Zooprophylactic Institute of Abruzzo and Molise "G. Caporale", Teramo, Italy
| | - Nobuhiro Yuki
- Department of Medicine, National University of Singapore, Singapore; Department of Physiology, National University of Singapore, Singapore
| | | | - Davide Corti
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Eduardo Nobile-Orazio
- 2nd Neurology, Department of Medical Biotechnology and Translational Medicine, IRCCS Humanitas Clinical Institute, Milan University, Rozzano, Milan, Italy
| | | | | | - Antonino Uncini
- Department of Neuroscience and Imaging, University "G. d'Annunzio", Chieti-Pescara, Italy
| |
Collapse
|
32
|
Peschl P, Reindl M, Schanda K, Sospedra M, Martin R, Lutterotti A. Antibody responses following induction of antigen-specific tolerance with antigen-coupled cells. Mult Scler 2014; 21:651-5. [PMID: 25200502 DOI: 10.1177/1352458514549405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have recently demonstrated the safety and tolerability of a novel therapeutic regimen employing autologous blood cells chemically coupled with seven myelin peptides to induce antigen-specific tolerance in MS (ETIMS study). The aim of the current study was an extended safety analysis to assess the effect of the ETIMS approach on antibodies to common autoantigens, the myelin peptides used and common recall antigens. None of the patients showed induction of autoantibody responses. One patient had a measurable myelin peptide-specific response at baseline, which was reduced after treatment. Total immunoglobulins and recall antibody responses showed no significant change.
Collapse
Affiliation(s)
- Patrick Peschl
- Clinical Department of Neurology, Innsbruck Medical University, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Austria
| | - Kathrin Schanda
- Clinical Department of Neurology, Innsbruck Medical University, Austria
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zurich, Switzerland
| | - Andreas Lutterotti
- Clinical Department of Neurology, Innsbruck Medical University, Austria/Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zurich, Switzerland
| |
Collapse
|
33
|
Affiliation(s)
- Lawrence Steinman
- Departments of Pediatrics, Neurology and Neurological Sciences, Stanford University, Stanford, California 94305;
| |
Collapse
|
34
|
Wang D, Bhat R, Sobel RA, Huang W, Wang LX, Olsson T, Steinman L. Uncovering cryptic glycan markers in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). Drug Dev Res 2014; 75:172-88. [PMID: 24648292 DOI: 10.1002/ddr.21169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/10/2014] [Indexed: 01/18/2023]
Abstract
Using an integrated antigen microarray approach, we observed epitope-spreading of autoantibody responses to a variety of antigenic structures in the cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS) and in the serum of mice with experimental autoimmune encephalomyelitis (EAE). These included previously described protein- and lipid-based antigenic targets and newly discovered autoimmunogenic sugar moieties, notably, autoantibodies specific for the oligomannoses in both MS patient CSF and the sera of mice with EAE. These glycans are often masked by other sugar moieties and belong to a class of cryptic autoantigens. We further determined that these targets are highly expressed on multiple cell types in MS and EAE lesions. Co-immunization of SJL/J mice with a Man9-KLH conjugate at the time of EAE induction elicited highly significant levels of anti-Man9-cluster autoantibodies. Nevertheless, this anti-glycan autoantibody response was associated with a significantly reduced clinical severity of EAE. The potential of these cryptic glycan markers and targeting antibodies for diagnostic and therapeutic interventions of neurological disorders has yet to be explored.
Collapse
Affiliation(s)
- Denong Wang
- Tumor Glycomics Laboratory, SRI International Biosciences Division, Menlo Park, CA, 94025, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Lim MJ, Liu Z, Braunschweiger KI, Awad A, Rothschild KJ. Correlated matrix-assisted laser desorption/ionization mass spectrometry and fluorescent imaging of photocleavable peptide-coded random bead-arrays. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2014; 28:49-62. [PMID: 24285390 PMCID: PMC3894740 DOI: 10.1002/rcm.6754] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/27/2013] [Accepted: 09/29/2013] [Indexed: 05/13/2023]
Abstract
RATIONALE Rapidly performing global proteomic screens is an important goal in the post-genomic era. Correlated matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) and fluorescent imaging of photocleavable peptide-coded random bead-arrays was evaluated as a critical step in a new method for proteomic screening that combines many of the advantages of MS with fluorescence-based microarrays. METHODS Small peptide-coded model bead libraries containing up to 20 different bead species were constructed by attaching peptides to 30-34 µm diameter glass, agarose or TentaGel® beads using photocleavable biotin or a custom-designed photocleavable linker. The peptide-coded bead libraries were randomly arrayed into custom gold-coated micro-well plates with 45 µm diameter wells and subjected to fluorescence and MALDI mass spectrometric imaging (MALDI-MSI). RESULTS Photocleavable mass-tags from individual beads in these libraries were spatially localized as ~65 µm spots using MALDI-MSI with high sensitivity and mass resolution. Fluorescently tagged beads were identified and correlated with their matching photocleavable mass-tags by comparing the fluorescence and MALDI-MS images of the same bead-array. Post-translational modification of the peptide Kemptide was also detected on individual beads in a photocleavable peptide-coded bead-array by MALDI-MSI alone, after exposure of the beads to protein kinase A (PKA). CONCLUSIONS Correlated MALDI-MS and fluorescent imaging of photocleavable peptide-coded random bead-arrays can provide a basis for performing global proteomic screening.
Collapse
Affiliation(s)
- Mark J Lim
- AmberGen, Incorporated313 Pleasant Street, Watertown, MA, 02472, USA
- * Correspondence to: M. J. Lim, AmberGen, Incorporated, 313 Pleasant Street, Watertown, MA 02472, USA., E-mail:
| | - Ziying Liu
- AmberGen, Incorporated313 Pleasant Street, Watertown, MA, 02472, USA
| | | | - Amany Awad
- AmberGen, Incorporated313 Pleasant Street, Watertown, MA, 02472, USA
| | - Kenneth J Rothschild
- AmberGen, Incorporated313 Pleasant Street, Watertown, MA, 02472, USA
- Molecular Biophysics Laboratory, Department of Physics and Photonics Center, Boston UniversityBoston, MA, 02215, USA
| |
Collapse
|
36
|
Schreiner B, Bailey SL, Miller SD. T-cell response dynamics in animal models of multiple sclerosis: implications for immunotherapies. Expert Rev Clin Immunol 2014; 3:57-72. [DOI: 10.1586/1744666x.3.1.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
37
|
Lolli F, Rovero P, Chelli M, Papini AM. Toward biomarkers in multiple sclerosis: new advances. Expert Rev Neurother 2014; 6:781-94. [PMID: 16734525 DOI: 10.1586/14737175.6.5.781] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis is an autoimmune disease that commonly affects young adults. If initially characterized by acute relapses, it is later followed by only incomplete remission. Over years, progressive disability and irreversible deficit lead to chronic neurological deficits in the majority of patients. The clinical course is protracted and unpredictable, and no biological marker is useful in predicting the evolution of autoaggression and disability. It is difficult to diagnose and to monitor disease progression after the initial symptoms or even during the major clinical manifestations, and it is difficult to treat. In this review, the authors report recent advances in the field, focusing on the search of new antigens as a marker of the disease, in their relevance to the pathophysiology and diagnosis of the disease.
Collapse
Affiliation(s)
- Francesco Lolli
- Laboratorio Interdipartimentale di Chimica & Biologia dei Peptidi & Proteine, Polo Scientifico e Tecnologico, Università degli Studi di Firenze, via Ugo Schiff 6, I-50019 Sesto Fiorentino, Italy.
| | | | | | | |
Collapse
|
38
|
Bansal AK. Role of bioinformatics in the development of new antibacterial therapy. Expert Rev Anti Infect Ther 2014; 6:51-65. [DOI: 10.1586/14787210.6.1.51] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Silva CL, Bonato VLD, dos Santos-Júnior RR, Zárate-Bladés CR, Sartori A. Recent advances in DNA vaccines for autoimmune diseases. Expert Rev Vaccines 2014; 8:239-52. [DOI: 10.1586/14760584.8.2.239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Gottlieb P, Utz PJ, Robinson W, Steinman L. Clinical optimization of antigen specific modulation of type 1 diabetes with the plasmid DNA platform. Clin Immunol 2013; 149:297-306. [PMID: 24094739 DOI: 10.1016/j.clim.2013.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 08/08/2013] [Indexed: 12/16/2022]
Abstract
Some clinical trials in humans have aimed at modulation of type 1 diabetes (T1D) via alteration of the immune response to putative islet cell antigens, particularly proinsulin and insulin, glutamic acid decarboxylase and the peptide, DiaPep 277, derived from heat shock protein 60. The focus here is on development of a specially engineered DNA plasmid encoding proinsulin to treat T1D. The plasmid is engineered to turn off adaptive immunity to proinsulin. This approach yielded exciting results in a randomized placebo controlled trial in 80 adult patients with T1D. The implications of this trial are explored in regards to the potential for sparing inflammation in islets and thus allowing the functioning beta cells to recover and produce more insulin. Strategies to further strengthen the effects seen thus far with the tolerizing DNA plasmid to proinsulin will be elucidated. The DNA platform affords an opportunity for easy modifications. In addition standard exploration of dose levels, route of administration and frequency of dose are practical. Optimization of the effects seen to date on C-peptide and on depletion of proinsulin specific CD8 T cells are feasible, with expected concomitant improvement in other parameters like hemoglobin A1c and reduction in insulin usage. T1D is one of the few autoimmune conditions where antigen specific therapy can be achieved, provided the approach is tested intelligently. Tolerizing DNA vaccines to proinsulin and other islet cell autoantigens is a worthy pursuit to potentially treat, prevent and to perhaps even 'cure' or 'prevent' type 1 diabetes.
Collapse
Affiliation(s)
- Peter Gottlieb
- Barbara Davis Center for Childhood Diabetes, Aurora, CO 80045-6511, USA
| | | | | | | |
Collapse
|
41
|
Mascanfroni ID, Yeste A, Vieira SM, Burns EJ, Patel B, Sloma I, Wu Y, Mayo L, Ben-Hamo R, Efroni S, Kuchroo VK, Robson SC, Quintana FJ. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat Immunol 2013; 14:1054-63. [PMID: 23995234 DOI: 10.1038/ni.2695] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 07/19/2013] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.
Collapse
Affiliation(s)
- Ivan D Mascanfroni
- 1] Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2]
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Van Haren K, Tomooka BH, Kidd BA, Banwell B, Bar-Or A, Chitnis T, Tenembaum SN, Pohl D, Rostasy K, Dale RC, O'Connor KC, Hafler DA, Steinman L, Robinson WH. Serum autoantibodies to myelin peptides distinguish acute disseminated encephalomyelitis from relapsing-remitting multiple sclerosis. Mult Scler 2013; 19:1726-33. [PMID: 23612879 DOI: 10.1177/1352458513485653] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Acute disseminated encephalomyelitis (ADEM) and relapsing-remitting multiple sclerosis (RRMS) share overlapping clinical, radiologic and laboratory features at onset. Because autoantibodies may contribute to the pathogenesis of both diseases, we sought to identify autoantibody biomarkers that are capable of distinguishing them. METHODS We used custom antigen arrays to profile anti-myelin-peptide autoantibodies in sera derived from individuals with pediatric ADEM (n = 15), pediatric multiple sclerosis (Ped MS; n = 11) and adult MS (n = 15). Using isotype-specific secondary antibodies, we profiled both IgG and IgM reactivities. We used Statistical Analysis of Microarrays software to confirm the differences in autoantibody reactivity profiles between ADEM and MS samples. We used Prediction Analysis of Microarrays software to generate and validate prediction algorithms, based on the autoantibody reactivity profiles. RESULTS ADEM was characterized by IgG autoantibodies targeting epitopes derived from myelin basic protein, proteolipid protein, myelin-associated oligodendrocyte basic glycoprotein, and alpha-B-crystallin. In contrast, MS was characterized by IgM autoantibodies targeting myelin basic protein, proteolipid protein, myelin-associated oligodendrocyte basic glycoprotein and oligodendrocyte-specific protein. We generated and validated prediction algorithms that distinguish ADEM serum (sensitivity 62-86%; specificity 56-79%) from MS serum (sensitivity 40-87%; specificity 62-86%) on the basis of combined IgG and IgM anti-myelin autoantibody reactivity to a small number of myelin peptides. CONCLUSIONS Combined profiles of serum IgG and IgM autoantibodies identified myelin antigens that may be useful for distinguishing MS from ADEM. Further studies are required to establish clinical utility. Further biological assays are required to delineate the pathogenic potential of these antibodies.
Collapse
Affiliation(s)
- Keith Van Haren
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
BACKGROUND The immune response involves the activation of heterogeneous populations of T cells and B cells that show different degrees of affinity and specificity for target antigens. Although several techniques have been developed to study the molecular pathways that control immunity, there is a need for high-throughput assays to monitor the specificity of the immune response. CONTENT Antigen microarrays provide a new tool to study the immune response. We reviewed the literature on antigen microarrays and their advantages and limitations, and we evaluated their use for the study of autoimmune diseases. Antigen arrays have been successfully used for several purposes in the investigation of autoimmune disorders: for disease diagnosis, to monitor disease progression and response to therapy, to discover mechanisms of pathogenesis, and to tailor antigen-specific therapies to the autoimmune response of individual patients. In this review we discuss the use of antigen microarrays for the study of 4 common autoimmune diseases and their animal models: type 1 diabetes, systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis. CONCLUSIONS Antigen microarrays constitute a new tool for the investigation of the immune response in autoimmune disorders and also in other conditions such as tumors and allergies. Once current limitations are overcome, antigen microarrays have the potential to revolutionize the investigation and management of autoimmune diseases.
Collapse
Affiliation(s)
- Ada Yeste
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
44
|
Robinson WH, Lindstrom TM, Cheung RK, Sokolove J. Mechanistic biomarkers for clinical decision making in rheumatic diseases. Nat Rev Rheumatol 2013; 9:267-76. [PMID: 23419428 DOI: 10.1038/nrrheum.2013.14] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The use of biomarkers is becoming increasingly intrinsic to the practice of medicine and holds great promise for transforming the practice of rheumatology. Biomarkers have the potential to aid clinical diagnosis when symptoms are present or to provide a means of detecting early signs of disease when they are not. Some biomarkers can serve as early surrogates of eventual clinical outcomes or guide therapeutic decision making by enabling identification of individuals likely to respond to a specific therapy. Using biomarkers might reduce the costs of drug development by enabling individuals most likely to respond to be enrolled in clinical trials, thereby minimizing the number of participants required. In this Review, we discuss the current use and the potential of biomarkers in rheumatology and in select fields at the forefront of biomarker research. We emphasize the value of different types of biomarkers, addressing the concept of 'actionable' biomarkers, which can be used to guide clinical decision making, and 'mechanistic' biomarkers, a subtype of actionable biomarker that is embedded in disease pathogenesis and, therefore, represents a potentially superior biomarker. We provide examples of actionable and mechanistic biomarkers currently available, and discuss how development of such biomarkers could revolutionize clinical practice and drug development.
Collapse
Affiliation(s)
- William H Robinson
- VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
45
|
On silico peptide microarrays for high-resolution mapping of antibody epitopes and diverse protein-protein interactions. Nat Med 2012; 18:1434-40. [PMID: 22902875 PMCID: PMC3491111 DOI: 10.1038/nm.2913] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 12/02/2011] [Indexed: 12/15/2022]
Abstract
We have developed a novel, silicon-based peptide array for broad biological applications, including potential for development as a real-time point-of-care platform. We employed photolithography on silicon wafers to synthesize microarrays (Intel arrays), containing every possible overlapping peptide within a linear protein sequence covering the N-terminal tail of human histone H2B. Arrays also included peptides with acetylated and methylated lysine residues reflecting post-translational modifications of H2B. We defined minimum binding epitopes for commercial antibodies recognizing modified and unmodified H2B peptides. We further demonstrated that this platform is suitable for highly sensitive methyltransferase and kinase substrate characterization. Intel arrays also revealed specific H2B epitopes recognized by autoantibodies in individuals with systemic lupus erythematosus (SLE) that have increased disease severity. By combining emerging nonfluorescence-based detection methods with an underlying integrated circuit, we are now poised to create a truly transformative proteomics platform with applications in bioscience, drug development, and clinical diagnostics.
Collapse
|
46
|
Devaux JJ. Antibodies to gliomedin cause peripheral demyelinating neuropathy and the dismantling of the nodes of Ranvier. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1402-13. [PMID: 22885108 DOI: 10.1016/j.ajpath.2012.06.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 06/15/2012] [Accepted: 06/20/2012] [Indexed: 11/18/2022]
Abstract
Guillain-Barré syndrome (GBS) and chronic inflammatory demyelinating polyneuropathy (CIDP) are conditions that affect peripheral nerves. The mechanisms that underlie demyelination in these neuropathies are unknown. Recently, we demonstrated that the node of Ranvier is the primary site of the immune attack in patients with GBS and CIDP. In particular, GBS patients have antibodies against gliomedin and neurofascin, two adhesion molecules that play a crucial role in the formation of nodes of Ranvier. We demonstrate that immunity toward gliomedin, but not neurofascin, induced a progressive neuropathy in Lewis rats characterized by conduction defects and demyelination in spinal nerves. The clinical symptoms closely followed the titers of anti-gliomedin IgG and were associated with an important deposition of IgG at nodes. Furthermore, passive transfer of antigliomedin IgG induced a severe demyelinating condition and conduction loss. In both active and passive models, the immune attack at nodes occasioned the loss of the nodal clusters for gliomedin, neurofascin-186, and voltage-gated sodium channels. These results indicate that primary immune reaction against gliomedin, a peripheral nervous system adhesion molecule, can be responsible for the initiation or progression of the demyelinating form of GBS. Furthermore, these autoantibodies affect saltatory propagation by dismantling nodal organization and sodium channel clusters. Antibodies reactive against nodal adhesion molecules thus likely participate in the pathologic process of GBS and CIDP.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Cell Adhesion Molecules, Neuronal/immunology
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Humans
- Immunity/immunology
- Immunization
- Immunization, Passive
- Immunoglobulin G/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Neuritis, Autoimmune, Experimental/immunology
- Neuritis, Autoimmune, Experimental/pathology
- Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology
- Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/pathology
- Ranvier's Nodes/immunology
- Ranvier's Nodes/pathology
- Rats
- Rats, Inbred Lew
- Spinal Nerve Roots/pathology
Collapse
Affiliation(s)
- Jérôme J Devaux
- National Center for Scientific Research (CNRS), Aix-Marseille University, Marseille, France.
| |
Collapse
|
47
|
Quintana FJ, Yeste A, Weiner HL, Covacu R. Lipids and lipid-reactive antibodies as biomarkers for multiple sclerosis. J Neuroimmunol 2012; 248:53-7. [PMID: 22579051 PMCID: PMC3667705 DOI: 10.1016/j.jneuroim.2012.01.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that targets the central nervous system (CNS). MS initially follows a relapsing-remitting course (RRMS) in which acute attacks are followed by a complete recovery. Eventually, 65% of the RRMS patients go on to develop secondary progressive MS (SPMS), characterized by the progressive and irreversible accumulation of neurological disability. It has been proposed that the transition from RRMS to SPMS results from changes in the nature of the inflammatory response and the progressive accumulation of neurodegeneration. To date, however, there is no reliable method to monitor the activity of the different immune and neurodegenerative processes that contribute to MS pathology. Thus, there is a need for biomarkers useful for the diagnosis, treatment and monitoring of MS patients. In this review, we discuss the potential use of lipids and the immune response against them as biomarkers of inflammation and neurodegeneration for MS.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
48
|
Nanoparticle-mediated codelivery of myelin antigen and a tolerogenic small molecule suppresses experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2012; 109:11270-5. [PMID: 22745170 DOI: 10.1073/pnas.1120611109] [Citation(s) in RCA: 243] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The immune response is normally controlled by regulatory T cells (Tregs). However, Treg deficits are found in autoimmune diseases, and therefore the induction of functional Tregs is considered a potential therapeutic approach for autoimmune disorders. The activation of the ligand-activated transcription factor aryl hydrocarbon receptor by 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) or other ligands induces dendritic cells (DCs) that promote FoxP3(+) Treg differentiation. Here we report the use of nanoparticles (NPs) to coadminister ITE and a T-cell epitope from myelin oligodendrocyte glycoprotein (MOG)(35)(-55) to promote the generation of Tregs by DCs. NP-treated DCs displayed a tolerogenic phenotype and promoted the differentiation of Tregs in vitro. Moreover, NPs carrying ITE and MOG(35-55) expanded the FoxP3(+) Treg compartment and suppressed the development of experimental autoimmune encephalomyelitis, an experimental model of multiple sclerosis. Thus, NPs are potential new tools to induce functional Tregs in autoimmune disorders.
Collapse
|
49
|
Abstract
Rheumatologists see patients with a range of autoimmune diseases. Phenotyping these diseases for diagnosis, prognosis and selection of therapies is an ever increasing problem. Advances in multiplexed assay technology at the gene, protein, and cellular level have enabled the identification of 'actionable biomarkers'; that is, biological metrics that can inform clinical practice. Not only will such biomarkers yield insight into the development, remission, and exacerbation of a disease, they will undoubtedly improve diagnostic sensitivity and accuracy of classification, and ultimately guide treatment. This Review provides an introduction to these powerful technologies that could promote the identification of actionable biomarkers, including mass cytometry, protein arrays, and immunoglobulin and T-cell receptor high-throughput sequencing. In our opinion, these technologies should become part of routine clinical practice for the management of autoimmune diseases. The use of analytical tools to deconvolve the data obtained from use of these technologies is also presented here. These analyses are revealing a more comprehensive and interconnected view of the immune system than ever before and should have an important role in directing future treatment approaches for autoimmune diseases.
Collapse
|
50
|
Sokolove J, Bromberg R, Deane KD, Lahey LJ, Derber LA, Chandra PE, Edison JD, Gilliland WR, Tibshirani RJ, Norris JM, Holers VM, Robinson WH. Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. PLoS One 2012; 7:e35296. [PMID: 22662108 PMCID: PMC3360701 DOI: 10.1371/journal.pone.0035296] [Citation(s) in RCA: 370] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 03/13/2012] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a prototypical autoimmune arthritis affecting nearly 1% of the world population and is a significant cause of worldwide disability. Though prior studies have demonstrated the appearance of RA-related autoantibodies years before the onset of clinical RA, the pattern of immunologic events preceding the development of RA remains unclear. To characterize the evolution of the autoantibody response in the preclinical phase of RA, we used a novel multiplex autoantigen array to evaluate development of the anti-citrullinated protein antibodies (ACPA) and to determine if epitope spread correlates with rise in serum cytokines and imminent onset of clinical RA. To do so, we utilized a cohort of 81 patients with clinical RA for whom stored serum was available from 1–12 years prior to disease onset. We evaluated the accumulation of ACPA subtypes over time and correlated this accumulation with elevations in serum cytokines. We then used logistic regression to identify a profile of biomarkers which predicts the imminent onset of clinical RA (defined as within 2 years of testing). We observed a time-dependent expansion of ACPA specificity with the number of ACPA subtypes. At the earliest timepoints, we found autoantibodies targeting several innate immune ligands including citrullinated histones, fibrinogen, and biglycan, thus providing insights into the earliest autoantigen targets and potential mechanisms underlying the onset and development of autoimmunity in RA. Additionally, expansion of the ACPA response strongly predicted elevations in many inflammatory cytokines including TNF-α, IL-6, IL-12p70, and IFN-γ. Thus, we observe that the preclinical phase of RA is characterized by an accumulation of multiple autoantibody specificities reflecting the process of epitope spread. Epitope expansion is closely correlated with the appearance of preclinical inflammation, and we identify a biomarker profile including autoantibodies and cytokines which predicts the imminent onset of clinical arthritis.
Collapse
Affiliation(s)
- Jeremy Sokolove
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
- * E-mail: (JS); (WHR)
| | - Reuven Bromberg
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Lauren J. Lahey
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Statistics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lezlie A. Derber
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Piyanka E. Chandra
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Statistics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jess D. Edison
- Walter Reed Army Medical Center, Washington, D.C., United States of America
| | | | - Robert J. Tibshirani
- Department of Statistics, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado, United States of America
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - William H. Robinson
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
- * E-mail: (JS); (WHR)
| |
Collapse
|