1
|
Ahsan S, Jafarpour S, Khoshnood MM, Nagesh D, Ho E, Ahsan N, Santoro JD. Anti-CD20 Therapy in Children With Severe Epstein-Barr Virus-Associated Meningoencephalitis. J Child Neurol 2025; 40:61-66. [PMID: 39234699 DOI: 10.1177/08830738241276972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Epstein-Barr virus meningoencephalitis is a rare central nervous system infection that lacks standardized treatment. Immunocompetent and immunosuppressed individuals with this condition frequently have poor prognostic outcomes, making the need to identify therapeutic interventions high. Here, we report 2 pediatric cases of severe Epstein-Barr virus meningoencephalitis, both unresponsive to immunoglobulin and corticosteroid therapy, who demonstrated rapid clinical recovery following rituximab administration. Prognostic outcomes revealed marked improvements in symptoms, neurologic function, and quality of life. Rituximab may offer therapeutic potential in severe and refractory Epstein-Barr virus meningoencephalitis through the medication's target of Epstein-Barr virus harboring B cells. This report emphasizes the need for timely evaluation and consideration of rituximab therapy in immunocompetent pediatric patients with Epstein-Barr virus meningoencephalitis.
Collapse
Affiliation(s)
- Sana Ahsan
- California University of Science and Medicine, Colton, CA, USA
| | - Saba Jafarpour
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Mellad M Khoshnood
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Deepti Nagesh
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Eugenia Ho
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Nusrat Ahsan
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Jonathan D Santoro
- Division of Neurology, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Kizer K, Augusto DG, Tubati A, Gomez R, Fouassier C, Gerungan C, Caspar CM, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Sabatino JJ, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. An autoantibody signature predictive for multiple sclerosis. Nat Med 2024; 30:1300-1308. [PMID: 38641750 DOI: 10.1038/s41591-024-02938-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. In this study, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster in approximately 10% of PwMS who share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active preclinical period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically or radiologically isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R Zamecnik
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gavin M Sowa
- University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ravi Dandekar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca D Bair
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kristen J Wade
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher M Bartley
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kerry Kizer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Danillo G Augusto
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Asritha Tubati
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Refujia Gomez
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Camille Fouassier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chloe Gerungan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Colette M Caspar
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Alexander
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anne E Wapniarski
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rita P Loudermilk
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Erica L Eggers
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kelsey C Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nora Jabassini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sabrina A Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Nicholas R Ragan
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Santaniello
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sergio E Baranzini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sabatino
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Riley M Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chu-Yueh Guo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey M Gelfand
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Richard Cuneo
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - H-Christian von Büdingen
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jill A Hollenbach
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Ari J Green
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen L Hauser
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mitchell T Wallin
- Department of Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC, USA
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Michael R Wilson
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Martinovic T, Vidicevic S, Ciric D, Bumbasirevic V, Stanojevic Z, Tasic J, Petricevic S, Isakovic A, Martinovic VC, Drndarevic N, Trajkovic V, Kravic-Stevovic T. The presence of Mott cells in the lymph nodes of rats with experimental autoimmune encephalomyelitis. Histochem Cell Biol 2024; 161:287-295. [PMID: 37952208 DOI: 10.1007/s00418-023-02252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Mott cells are plasma cells that have multiple spherical Russell bodies packed in their cytoplasm. Russell bodies are dilated endoplasmic reticulum cisternae filled with aggregates of immunoglobulins that are neither secreted nor degraded. Mott cells were observed in our study by light and electron microscope in the lymph nodes of rats with experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Mott cells were detected on hematoxylin and eosin (HE)-stained lymph node sections as vacuolated cells with eccentrically positioned nuclei and large number of faint blue spherical inclusions in the cytoplasm. Electron microscopic investigation revealed the presence of Russell bodies of the "medusa" form inside Mott cells in lymph node ultra-thin sections of EAE animals. Mott cells expressed the plasma cell marker CD138 and either kappa or lambda immunoglobulin light chains, indicating their origin from polyclonally activated B cells. Finally, Mott cells were associated with active EAE, as they were not found in the lymph nodes of EAE-resistant Albino Oxford rats. The presence of Russell bodies implies an excessive production of immunoglobulins in EAE, thus further emphasizing the role of B cells, and among them Mott cells, in the pathogenesis of this animal model of multiple sclerosis.
Collapse
Affiliation(s)
- Tamara Martinovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia
| | - Sasenka Vidicevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Darko Ciric
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia.
| | - Vladimir Bumbasirevic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Zeljka Stanojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Tasic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Sasa Petricevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Isakovic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tamara Kravic-Stevovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000, Belgrade, Serbia
| |
Collapse
|
4
|
Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Tubati A, Gomez R, Fouassier C, Gerungan C, Alexander J, Wapniarski AE, Loudermilk RP, Eggers EL, Zorn KC, Ananth K, Jabassini N, Mann SA, Ragan NR, Santaniello A, Henry RG, Baranzini SE, Zamvil SS, Bove RM, Guo CY, Gelfand JM, Cuneo R, von Büdingen HC, Oksenberg JR, Cree BAC, Hollenbach JA, Green AJ, Hauser SL, Wallin MT, DeRisi JL, Wilson MR. A Predictive Autoantibody Signature in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.01.23288943. [PMID: 37205595 PMCID: PMC10187343 DOI: 10.1101/2023.05.01.23288943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Although B cells are implicated in multiple sclerosis (MS) pathophysiology, a predictive or diagnostic autoantibody remains elusive. Here, the Department of Defense Serum Repository (DoDSR), a cohort of over 10 million individuals, was used to generate whole-proteome autoantibody profiles of hundreds of patients with MS (PwMS) years before and subsequently after MS onset. This analysis defines a unique cluster of PwMS that share an autoantibody signature against a common motif that has similarity with many human pathogens. These patients exhibit antibody reactivity years before developing MS symptoms and have higher levels of serum neurofilament light (sNfL) compared to other PwMS. Furthermore, this profile is preserved over time, providing molecular evidence for an immunologically active prodromal period years before clinical onset. This autoantibody reactivity was validated in samples from a separate incident MS cohort in both cerebrospinal fluid (CSF) and serum, where it is highly specific for patients eventually diagnosed with MS. This signature is a starting point for further immunological characterization of this MS patient subset and may be clinically useful as an antigen-specific biomarker for high-risk patients with clinically- or radiologically-isolated neuroinflammatory syndromes.
Collapse
Affiliation(s)
- Colin R. Zamecnik
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Gavin M. Sowa
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Ravi Dandekar
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rebecca D. Bair
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kristen J. Wade
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Christopher M. Bartley
- UCSF Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Asritha Tubati
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Refujia Gomez
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Camille Fouassier
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chloe Gerungan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jessica Alexander
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne E. Wapniarski
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Rita P. Loudermilk
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Erica L. Eggers
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Kelsey C. Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sabrina A. Mann
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Nicholas R. Ragan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Adam Santaniello
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Roland G. Henry
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Sergio E. Baranzini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Scott S. Zamvil
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Riley M. Bove
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Chu-Yueh Guo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jeffrey M. Gelfand
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Richard Cuneo
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - H.-Christian von Büdingen
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jorge R. Oksenberg
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce AC Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Jill A. Hollenbach
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA USA
| | - Ari J. Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Stephen L. Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Mitchell T. Wallin
- Veterans Affairs, Multiple Sclerosis Center of Excellence, Washington, DC and University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Michael R. Wilson
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
5
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
6
|
Central role of B cells in interleukin-23 dependent neuroinflammation in the GF-IL23 model. Neuroreport 2022; 33:577-582. [DOI: 10.1097/wnr.0000000000001818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Skewing of the Antibody Repertoire in Cerebrospinal Fluid B Cells from Healthy Controls and Patients with Schizophrenia. Behav Brain Res 2022; 422:113743. [PMID: 35007628 PMCID: PMC9248756 DOI: 10.1016/j.bbr.2022.113743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/20/2022]
Abstract
Autoantibodies play a role in the etiology of some neuropsychiatric disorders. To address the possibility that B cells and their antibodies may be involved in the pathophysiology of schizophrenia, we examined B cells in cerebrospinal fluid (CSF) and peripheral blood (PB) of 4 schizophrenic patients (SP) and 4 healthy control (HC) volunteers by analyzing immunoglobulin VH gene usage. All CSF samples contained measurable levels of B cells. We found for both SP and HC, CSF B cells represented a select subset of, and were not the same as, B cells in PB. Moreover, we found statistically significant differences in antibodies generated by CSF B cells in SP compared to CSF B cells in HC. Although binding characteristics of CSF SP-associated B cell antibodies is unknown, the study number is small, and pathophysiology has not been established, these results suggest the value of focusing further study on the distinctly separate population of CSF B cells in SP.
Collapse
|
8
|
Kim S, Ahn SJ, Chu K. Epstein-Barr virus-associated acute disseminated encephalomyelitis successfully treated with rituximab: a case report. ENCEPHALITIS 2021; 1:85-88. [DOI: 10.47936/encephalitis.2021.00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/03/2021] [Indexed: 02/05/2023] Open
|
9
|
Nitsch L, Petzinna S, Zimmermann J, Schneider L, Krauthausen M, Heneka MT, Getts DR, Becker A, Müller M. Astrocyte-specific expression of interleukin 23 leads to an aggravated phenotype and enhanced inflammatory response with B cell accumulation in the EAE model. J Neuroinflammation 2021; 18:101. [PMID: 33906683 PMCID: PMC8080359 DOI: 10.1186/s12974-021-02140-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/26/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Interleukin 23 is a critical cytokine in the pathogenesis of multiple sclerosis. But the local impact of interleukin 23 on the course of neuroinflammation is still not well defined. To further characterize the effect of interleukin 23 on CNS inflammation, we recently described a transgenic mouse model with astrocyte-specific expression of interleukin 23 (GF-IL23 mice). The GF-IL23 mice spontaneously develop a progressive ataxic phenotype with cerebellar tissue destruction and inflammatory infiltrates with high amounts of B cells most prominent in the subarachnoid and perivascular space. METHODS To further elucidate the local impact of the CNS-specific interleukin 23 synthesis in autoimmune neuroinflammation, we induced a MOG35-55 experimental autoimmune encephalomyelitis (EAE) in GF-IL23 mice and WT mice and analyzed the mice by histology, flow cytometry, and transcriptome analysis. RESULTS We were able to demonstrate that local interleukin 23 production in the CNS leads to aggravation and chronification of the EAE course with a severe paraparesis and an ataxic phenotype. Moreover, enhanced multilocular neuroinflammation was present not only in the spinal cord, but also in the forebrain, brainstem, and predominantly in the cerebellum accompanied by persisting demyelination. Thereby, interleukin 23 creates a pronounced proinflammatory response with accumulation of leukocytes, in particular B cells, CD4+ cells, but also γδ T cells and activated microglia/macrophages. Furthermore, transcriptome analysis revealed an enhanced proinflammatory cytokine milieu with upregulation of lymphocyte activation markers, co-stimulatory markers, chemokines, and components of the complement system. CONCLUSION Taken together, the GF-IL23 model allowed a further breakdown of the different mechanisms how IL-23 drives neuroinflammation in the EAE model and proved to be a useful tool to further dissect the impact of interleukin 23 on neuroinflammatory models.
Collapse
Affiliation(s)
- Louisa Nitsch
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.
| | - Simon Petzinna
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Julian Zimmermann
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Linda Schneider
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.,Department of Surgery, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Marius Krauthausen
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Daniel R Getts
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Albert Becker
- Department of Neuropathology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany
| | - Marcus Müller
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, D-53127, Bonn, Germany.,School of Molecular Bioscience, University of Sydney, Sydney, Australia
| |
Collapse
|
10
|
Anti-Kir4.1 Antibodies in Multiple Sclerosis: Specificity and Pathogenicity. Int J Mol Sci 2020; 21:ijms21249632. [PMID: 33348803 PMCID: PMC7765826 DOI: 10.3390/ijms21249632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/17/2022] Open
Abstract
The glial cells in the central nervous system express diverse inward rectifying potassium channels (Kir). They express multiple Kir channel subtypes that are likely to have distinct functional roles related to their differences in conductance, and sensitivity to intracellular and extracellular factors. Dysfunction in a major astrocyte potassium channel, Kir4.1, appears as an early pathological event underlying neuronal phenotypes in several neurological diseases. The autoimmune effects on the potassium channel have not yet been fully described in the literature. However, several research groups have reported that the potassium channels are an immune target in patients with various neurological disorders. In 2012, Srivastava et al. reported about Kir4.1, a new immune target for autoantibodies in patients with multiple sclerosis (MS). Follow-up studies have been conducted by several research groups, but no clear conclusion has been reached. Most follow-up studies, including ours, have reported that the prevalence of Kir4.1-seropositive patients with MS was lower than that in the initial study. Therefore, we extensively review studies on the method of antibody testing, seroprevalence of MS, and other neurological diseases in patients with MS. Finally, based on the role of Kir4.1 in MS, we consider whether it could be an immune target in this disease.
Collapse
|
11
|
Eslami A, Dehbashi M, Ashja-Arvan M, Salehi H, Azimzadeh M, Ganjalikhani-Hakemi M. Assessment of ability of human adipose derived stem cells for long term overexpression of IL-11 and IL-13 as therapeutic cytokines. Cytotechnology 2020; 72:773-784. [PMID: 32935166 PMCID: PMC7547926 DOI: 10.1007/s10616-020-00421-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/02/2020] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) are a type of mesenchymal stem cells with the therapeutic effects that make them one of the best sources for cell therapy. In this study, we aimed to assess the ability of human ADSCs for constant expression of IL-11 and IL-13, simultaneously. In this study, the characterized hADSCs were transduced with a lentiviral vector (PCDH-513B) containing IL-11 and IL-13 genes, and the ability of long-term expression of the transgenes was evaluated by ELISA technique on days 15, 45 and 75 after transduction. Our results indicated a high rate of transduction (more than 90%) in the isolated hADSCs. Our data showed the highest rate of expression on days 75 after transduction which was 242.67 pg/ml for IL-11 and 303.6 pg/ml for IL-13 compared with 35.2 pg/ml and 35.6 pg/ml in untreated cells, respectively (p = 0.001). Besides, MTT assay showed transduction of hADSCs with lentiviral viruses containing IL-11 and IL-13 had no adverse effect on hADSCs proliferation (p-value = 0.89). Finally, we successfully constructed a hADSC population stably overexpressing IL-11 as the neurotrophic cytokine and IL-13 as the anti-inflammatory cytokine and this transduced cells can be used for further studies in EAE mice model.
Collapse
Affiliation(s)
- Asma Eslami
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Cell and Molecular Biology, Faculty of Biological Sciences and Technologies, University of Isfahan, 81746-73441 Isfahan, Iran
| | - Mehnoosh Ashja-Arvan
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Azimzadeh
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
12
|
Klöß S, Dehmel S, Braun A, Parnham MJ, Köhl U, Schiffmann S. From Cancer to Immune-Mediated Diseases and Tolerance Induction: Lessons Learned From Immune Oncology and Classical Anti-cancer Treatment. Front Immunol 2020; 11:1423. [PMID: 32733473 PMCID: PMC7360838 DOI: 10.3389/fimmu.2020.01423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/02/2020] [Indexed: 12/27/2022] Open
Abstract
Success in cancer treatment over the last four decades has ranged from improvements in classical drug therapy to immune oncology. Anti-cancer drugs have also often proven beneficial for the treatment of inflammatory and autoimmune diseases. In this review, we report on challenging examples that bridge between treatment of cancer and immune-mediated diseases, addressing mechanisms and experimental models as well as clinical investigations. Patient-derived tumor xenograft (PDX) (humanized) mouse models represent useful tools for preclinical evaluation of new therapies and biomarker identification. However, new developments using human ex vivo approaches modeling cancer, for example in microfluidic human organs-on-chips, promise to identify key molecular, cellular and immunological features of human cancer progression in a fully human setting. Classical drugs which bridge the gap, for instance, include cytotoxic drugs, proteasome inhibitors, PI3K/mTOR inhibitors and metabolic inhibitors. Biologicals developed for cancer therapy have also shown efficacy in the treatment of autoimmune diseases. In immune oncology, redirected chimeric antigen receptor (CAR) T cells have achieved spectacular remissions in refractory B cell leukemia and lymphoma and are currently under development for tolerance induction using cell-based therapies such as CAR Tregs or NK cells. Finally, a brief outline will be given of the lessons learned from bridging cancer and autoimmune diseases as well as tolerance induction.
Collapse
Affiliation(s)
- Stephan Klöß
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School (MHH), Hanover, Germany
| | - Susann Dehmel
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Frankfurt, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School (MHH), Hanover, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Susanne Schiffmann
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch for Translational Medicine and Pharmacology (TMP), Frankfurt, Germany
| |
Collapse
|
13
|
Dhenni R, Phan TG. The geography of memory B cell reactivation in vaccine-induced immunity and in autoimmune disease relapses. Immunol Rev 2020; 296:62-86. [PMID: 32472583 DOI: 10.1111/imr.12862] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/05/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Memory B cells (Bmem) provide an active second layer of defense against re-infection by pathogens that have bypassed the passive first layer provided by neutralizing antibodies. Here, we review recent progress in our understanding of Bmem heterogeneity in terms of their origin (germinal center-dependent vs center-independent), phenotype (canonical vs atypical vs age-associated B cells), trafficking (recirculating vs tissue-resident), and fate (plasma cell vs germinal center differentiation). The development of transgenic models and intravital imaging technologies has made it possible to track the cellular dynamics of Bmem reactivation by antigen, their interactions with follicular memory T cells, and differentiation into plasma cells in subcapsular proliferative foci in the lymph nodes of immune animals. Such in situ studies have reinforced the importance of geography in shaping the outcome of the secondary antibody response. We also review the evidence for Bmem reactivation and differentiation into short-lived plasma cells in the pathogenesis of disease flares in relapsing-remitting autoimmune diseases. Elucidating the mechanisms that control the Bmem fate decision to differentiate into plasma cells or germinal center B cells will aid future efforts to more precisely engineer fit-for-purpose vaccines as well as to treat antibody-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Rama Dhenni
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
14
|
Kappa Free Light Chains and IgG Combined in a Novel Algorithm for the Detection of Multiple Sclerosis. Brain Sci 2020; 10:brainsci10060324. [PMID: 32471086 PMCID: PMC7349504 DOI: 10.3390/brainsci10060324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 01/09/2023] Open
Abstract
Background: It is well known that the cerebrospinal fluid (CSF) concentrations of free light chains (FLC) and immunoglobulin G (IgG) are elevated in multiple sclerosis patients (MS). Therefore, in this study we aimed to develop a model based on the concentrations of free light chains and IgG to predict multiple sclerosis. We tried to evaluate the diagnostic usefulness of the novel κIgG index and λIgG index, here presented for the first time, and compare them with the κFLC index and the λFLC index in multiple sclerosis patients. Methods: CSF and serum samples were obtained from 76 subjects who underwent lumbar puncture for diagnostic purposes and, as a result, were divided into two groups: patients with multiple sclerosis (n = 34) and patients with other neurological disorders (control group; n = 42). The samples were analyzed using turbidimetry and isoelectric focusing. The κIgG index, λIgG index, κFLC index, and λFLC index were calculated using specific formulas. Results: The concentrations of CSF κFLC, CSF λFLC, and serum κFLC and the values of κFLC index, λFLC index, and κIgG index were significantly higher in patients with multiple sclerosis compared to controls. CSF κFLC concentration and the values of κFLC index, λFLC index, and κIgG index differed in patients depending on their pattern type of oligoclonal bands. κFLC concentration was significantly higher in patients with pattern type 2 and type 3 in comparison to those with pattern type 1 and type 4. The κFLC index, λFLC index, and κIgG index were significantly higher in patients with pattern type 2 in comparison to those with pattern type 4. The κFLC index and κIgG index were significantly higher in patients with pattern type 2 in comparison to those with pattern type 1, and in patients with pattern type 3 compared to those with pattern type 4. The κIgG index was markedly elevated in patients with pattern type 3 compared to those with pattern type 1. In the total study group, κFLC, λFLC, κFLC index, λFLC index, κIgG index, and λIgG index correlated with each other. The κIgG index showed the highest diagnostic power (area under the curve, AUC) in the detection of multiple sclerosis. The κFLC index and κIgG index showed the highest diagnostic sensitivity, and the κIgG index presented the highest ability to exclude multiple sclerosis. Conclusion: This study provides novel information about the diagnostic significance of four markers combined in the κIgG index. More investigations in larger study groups are needed to confirm that the κIgG index can reflect the intrathecal synthesis of immunoglobulins and may improve the diagnosis of multiple sclerosis.
Collapse
|
15
|
Graner M, Pointon T, Manton S, Green M, Dennison K, Davis M, Braiotta G, Craft J, Edwards T, Polonsky B, Fringuello A, Vollmer T, Yu X. Oligoclonal IgG antibodies in multiple sclerosis target patient-specific peptides. PLoS One 2020; 15:e0228883. [PMID: 32084151 PMCID: PMC7034880 DOI: 10.1371/journal.pone.0228883] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/24/2020] [Indexed: 12/04/2022] Open
Abstract
IgG oligoclonal bands (OCBs) are present in the cerebrospinal fluid (CSF) of more than 95% of patients with multiple sclerosis (MS), and are considered to be the immunological hallmark of disease. However, the target specificities of the IgG in MS OCBs have remained undiscovered. Nevertheless, evidence that OCBs are associated with increased levels of disease activity and disability support their probable pathological role in MS. We investigated the antigen specificity of individual MS CSF IgG from 20 OCB-positive patients and identified 40 unique peptides by panning phage-displayed random peptide libraries. Utilizing our unique techniques of phage-mediated real-time Immuno-PCR and phage-probed isoelectric focusing immunoblots, we demonstrated that these peptides were targeted by intrathecal oligoclonal IgG antibodies of IgG1 and IgG3 subclasses. In addition, we showed that these peptides represent epitopes sharing sequence homologies with proteins of viral origin, and proteins involved in cell stress, apoptosis, and inflammatory processes. Although homologous peptides were found within individual patients, no shared peptide sequences were found among any of the 42 MS and 13 inflammatory CSF control specimens. The distinct sets of oligoclonal IgG-reactive peptides identified by individual MS CSF suggest that the elevated intrathecal antibodies may target patient-specific antigens.
Collapse
Affiliation(s)
- Michael Graner
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Tiffany Pointon
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Sean Manton
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Miyoko Green
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kathryn Dennison
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mollie Davis
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Gino Braiotta
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Julia Craft
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Taylor Edwards
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Bailey Polonsky
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Anthony Fringuello
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Timothy Vollmer
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Xiaoli Yu
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
16
|
Cavallo S. Immune-mediated genesis of multiple sclerosis. J Transl Autoimmun 2020; 3:100039. [PMID: 32743522 PMCID: PMC7388381 DOI: 10.1016/j.jtauto.2020.100039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is widely acknowledged to be an autoimmune disease affecting the neuronal myelin structure of the CNS. Autoantigens recognized as the target of this autoimmune process are: myelin basal protein, anti-proteolipid protein, antimyelin-associated glycoprotein and antimyelin-based oligodendrocytic basic protein. Ample evidence supports the idea of a dysregulation of immunological tolerance towards self-antigens of neuronal myelin structure triggered by one or more viral or bacterial microbial agents in predisposed HLA gene subjects. Genetic predisposition to MS has been highlighted by numerous studies associating the disease to specific HLA haplotypes. Moreover, a wide range of evidence supports the fact that MS may be consequence of one or more viral or bacterial infections such as measles virus, EBV, HHV6, HZV, Chlamydia pneumoniae, Helicobacter Pylori, and other microbial agents. Microbiota elements also seems to have a role on the determinism of the disease as a pathogenic or protective factor. The autoimmune pathogenetic process could arise when a molecular mimicry between a foreign microbial antigen and an auto-antigen occurs in an HLA gene subject competent for that particular antigen. The antigen-presenting cells in this case would induce the activation of a specific Th clone causing a cross-reaction between a foreign antigen and an autoantigen resulting in an autoimmune response. A multifactorial ethiopathogenetic model based on immunomediation is a reliable hypothesis for multiple sclerosis. Evidence found in the scientific literature makes it possible to reconstruct this etiopathogenetic hypothesis for MS. HLA gene predisposition, correlation with infections, molecular mimicry and other immunological data are reported.
Collapse
Affiliation(s)
- Salvatore Cavallo
- Expert Doctor in Non-Conventional Medicine, Professor and Member of the Board of the MMS, MMS (Medicina di Modulazione Dei Sistemi) Roma, Salvatore Cavallo Via G.B. Pergolesi, 28, 75100, Matera, Italy
| |
Collapse
|
17
|
Qiu K, He Q, Chen X, Liu H, Deng S, Lu W. Pregnancy-Related Immune Changes and Demyelinating Diseases of the Central Nervous System. Front Neurol 2019; 10:1070. [PMID: 31649614 PMCID: PMC6794637 DOI: 10.3389/fneur.2019.01070] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/23/2019] [Indexed: 12/31/2022] Open
Abstract
Demyelinating diseases of the central nervous system comprise a heterogeneous group of autoimmune disorders characterized by myelin loss with relative sparing of axons occurring on a background of inflammation. Some of the most common demyelinating diseases are multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Besides showing clinical, radiological, and histopathological features that complicate their diagnosis, demyelinating diseases often involve different immunological processes that produce distinct inflammatory patterns. Evidence of demyelination diseases derives mostly from animal studies of experimental autoimmune encephalomyelitis (EAE), a model that relies on direct antibody–antigen interactions induced by encephalitogenic T cells. Pregnancy is characterized by non-self-recognition, immunomodulatory changes and an altered Th1/Th2 balance, generally considered a Th2-type immunological state that protects the mother from infections. During pregnancy, the immune response of patients with autoimmune disease complicated with pregnancy is different. Immune tolerance in pregnancy may affect the course of some diseases, which may reach remission or be exacerbated. In this review, we summarize current knowledge on the immune status during pregnancy and discuss the relationship between pregnancy-related immune changes and demyelinating diseases of the central nervous system.
Collapse
Affiliation(s)
- Ke Qiu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiang He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiqian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuwen Deng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Nitsch L, Zimmermann J, Krauthausen M, Hofer MJ, Saggu R, Petzold GC, Heneka MT, Getts DR, Becker A, Campbell IL, Müller M. CNS-Specific Synthesis of Interleukin 23 Induces a Progressive Cerebellar Ataxia and the Accumulation of Both T and B Cells in the Brain: Characterization of a Novel Transgenic Mouse Model. Mol Neurobiol 2019; 56:7977-7993. [PMID: 31154574 DOI: 10.1007/s12035-019-1640-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022]
Abstract
Interleukin 23 (IL-23) is a key mediator in neuroinflammation in numerous autoimmune diseases including multiple sclerosis (MS). However, the pathophysiology of IL-23 and how it contributes to neuroinflammation is poorly defined. To further clarify the role of IL-23 in CNS inflammation, we generated a transgenic mouse model (GF-IL23) with astrocyte-targeted expression of both IL-23 subunits, IL-23p19, and IL-23p40. These GF-IL23 mice spontaneously develop a progressive ataxic phenotype, which corresponds to cerebellar tissue destruction, and inflammatory infiltrates most prominent in the subarachnoidal and perivascular space. The CNS-cytokine milieu was characterized by numerous inflammatory mediators such as IL-17a and IFNγ. However, the leukocytic infiltrates were surprisingly predominated by B cells. To further examine the impact of the CNS-specific IL-23 synthesis on an additional systemic inflammatory stimulus, we applied the LPS-induced endotoxemia model. Administration of LPS in GF-IL23 mice resulted in early and pronounced microglial activation, enhanced cytokine production and, in sharp contrast to control animals, in the formation of lymphocytic infiltrates. Our model confirms a critical role for IL-23 in the induction of inflammation in the CNS, in particular facilitating the accumulation of lymphocytes in and around the brain. Thereby, CNS-specific synthesis of IL-23 is able to induce a cascade of inflammatory cytokines leading to microglia activation, astrocytosis, and ultimately tissue damage. The presented transgenic model will be a useful tool to further dissect the role of IL-23 in neuroinflammation.
Collapse
Affiliation(s)
- Louisa Nitsch
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Julian Zimmermann
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Marius Krauthausen
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Markus J Hofer
- School of Life and Environmental Sciences, Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Raman Saggu
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, D-53127, Bonn, Germany
| | - Gabor C Petzold
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Str. 27, D-53127, Bonn, Germany
| | - Michael T Heneka
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
- Clinical Neuroscience Unit, Universitaetsklinikum Bonn, Bonn, Germany
| | - Daniel R Getts
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Albert Becker
- Department of Neuropathology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Iain L Campbell
- School of Molecular Bioscience, University of Sydney, Sydney, Australia
| | - Marcus Müller
- Department of Neurology, Universitaetsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany.
- School of Molecular Bioscience, University of Sydney, Sydney, Australia.
| |
Collapse
|
19
|
Van Kaer L, Postoak JL, Wang C, Yang G, Wu L. Innate, innate-like and adaptive lymphocytes in the pathogenesis of MS and EAE. Cell Mol Immunol 2019; 16:531-539. [PMID: 30874627 DOI: 10.1038/s41423-019-0221-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) in which the immune system damages the protective insulation surrounding the nerve fibers that project from neurons. A hallmark of MS and its animal model, experimental autoimmune encephalomyelitis (EAE), is autoimmunity against proteins of the myelin sheath. Most studies in this field have focused on the roles of CD4+ T lymphocytes, which form part of the adaptive immune system as both mediators and regulators in disease pathogenesis. Consequently, the treatments for MS often target the inflammatory CD4+ T-cell responses. However, many other lymphocyte subsets contribute to the pathophysiology of MS and EAE, and these subsets include CD8+ T cells and B cells of the adaptive immune system, lymphocytes of the innate immune system such as natural killer cells, and subsets of innate-like T and B lymphocytes such as γδ T cells, natural killer T cells, and mucosal-associated invariant T cells. Several of these lymphocyte subsets can act as mediators of CNS inflammation, whereas others exhibit immunoregulatory functions in disease. Importantly, the efficacy of some MS treatments might be mediated in part by effects on lymphocytes other than CD4+ T cells. Here we review the contributions of distinct subsets of lymphocytes on the pathogenesis of MS and EAE, with an emphasis on lymphocytes other than CD4+ T cells. A better understanding of the distinct lymphocyte subsets that contribute to the pathophysiology of MS and its experimental models will inform the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Joshua L Postoak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Chuan Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
20
|
Seraji-Bozorgzad N, Khan O, Cree BA, Bao F, Caon C, Zak I, Razmjou S, Tselis A, Millis S, Bernitsas E. Cerebral Gray Matter Atrophy Is Associated with the CSF IgG index in African American with Multiple Sclerosis. J Neuroimaging 2017; 27:476-480. [DOI: 10.1111/jon.12435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/09/2017] [Indexed: 02/01/2023] Open
Affiliation(s)
- Navid Seraji-Bozorgzad
- Sastry Foundation Advanced Imaging Laboratory, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Omar Khan
- Sastry Foundation Advanced Imaging Laboratory, Department of Neurology; Wayne State University School of Medicine; Detroit MI
- Multiple Sclerosis Center, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Bruce A.C. Cree
- Multiple Sclerosis Center, Department of Neurology; University of California; San Francisco CA
| | - Fen Bao
- Sastry Foundation Advanced Imaging Laboratory, Department of Neurology; Wayne State University School of Medicine; Detroit MI
- Multiple Sclerosis Center, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Christina Caon
- Multiple Sclerosis Center, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Imad Zak
- Department of Radiology; Wayne State University School of Medicine; Detroit MI
| | - Sara Razmjou
- Sastry Foundation Advanced Imaging Laboratory, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Alexandros Tselis
- Multiple Sclerosis Center, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| | - Scott Millis
- Division of Biostatistics, Department of Physical Medicine & Rehabilitation; Wayne State University School of Medicine; Detroit MI
| | - Evanthia Bernitsas
- Multiple Sclerosis Center, Department of Neurology; Wayne State University School of Medicine; Detroit MI
| |
Collapse
|
21
|
Casserly CS, Nantes JC, Whittaker Hawkins RF, Vallières L. Neutrophil perversion in demyelinating autoimmune diseases: Mechanisms to medicine. Autoimmun Rev 2017; 16:294-307. [PMID: 28161558 DOI: 10.1016/j.autrev.2017.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
|
22
|
Northrup L, Sullivan BP, Hartwell BL, Garza A, Berkland C. Screening Immunomodulators To Skew the Antigen-Specific Autoimmune Response. Mol Pharm 2016; 14:66-80. [PMID: 28043135 DOI: 10.1021/acs.molpharmaceut.6b00725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Current therapies to treat autoimmune diseases often result in side effects such as nonspecific immunosuppression. Therapies that can induce antigen-specific immune tolerance provide an opportunity to reverse autoimmunity and mitigate the risks associated with global immunosuppression. In an effort to induce antigen-specific immune tolerance, co-administration of immunomodulators with autoantigens has been investigated in an effort to reprogram autoimmunity. To date, identifying immunomodulators that may skew the antigen-specific immune response has been ad hoc at best. To address this need, we utilized splenocytes obtained from mice with experimental autoimmune encephalomyelitis (EAE) in order to determine if certain immunomodulators may induce markers of immune tolerance following antigen rechallenge. Of the immunomodulatory compounds investigated, only dexamethasone modified the antigen-specific immune response by skewing the cytokine response and decreasing T-cell populations at a concentration corresponding to a relevant in vivo dose. Thus, antigen-educated EAE splenocytes provide an ex vivo screen for investigating compounds capable of skewing the antigen-specific immune response, and this approach could be extrapolated to antigen-educated cells from other diseases or human tissues.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States
| | - Aaron Garza
- Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States.,Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States.,Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| |
Collapse
|
23
|
DNA Methylation: a New Player in Multiple Sclerosis. Mol Neurobiol 2016; 54:4049-4059. [PMID: 27314687 DOI: 10.1007/s12035-016-9966-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a neurological and chronic inflammatory disease that is mediated by demyelination and axonal degeneration in the central nervous system (CNS). Studies have shown that immune system components such as CD4+, CD8+, CD44+ T cells, B lymphatic cells, and inflammatory cytokines play a critical role in inflammatory processes and myelin damage associated with MS. Nevertheless, the pathogenesis of MS remains poorly defined. DNA methylation, a significant epigenetic modification, is reported to be extensively involved in MS pathogenesis through the regulation of gene expression. This review focuses on DNA methylation involved in MS pathogenesis. Evidence showed the hypermethylation of human leukocyte antigen-DRB1 (HLA-DRB1) in CD4+ T cells, the genome-wide DNA methylation in CD8+ T cells, the hypermethylation of interleukin-4 (IL-4)/forkhead winged helix transcription factor 3 (Foxp3), and the demethylation of interferon-γ (IFN-γ)/IL-17a in CD44+ encephalitogenic T cells. Studies also showed the hypermethylation of SH2-containing protein tyrosine phosphatase-1 (SHP-1) in peripheral blood mononuclear cells (PBMCs) and methylated changes of genes regulating oligodendrocyte and neuronal function in normal-appearing white matter. Clarifying the mechanism of aberrant methylation on MS may explain part of the pathology and will lead to the development of a new therapeutic target for the treatment of MS in the future.
Collapse
|
24
|
Willis SN, Stathopoulos P, Chastre A, Compton SD, Hafler DA, O'Connor KC. Investigating the Antigen Specificity of Multiple Sclerosis Central Nervous System-Derived Immunoglobulins. Front Immunol 2015; 6:600. [PMID: 26648933 PMCID: PMC4663633 DOI: 10.3389/fimmu.2015.00600] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/09/2015] [Indexed: 12/25/2022] Open
Abstract
The central nervous system (CNS) of patients with multiple sclerosis (MS) is the site where disease pathology is evident. Damaged CNS tissue is commonly associated with immune cell infiltration. This infiltrate often includes B cells that are found in multiple locations throughout the CNS, including the cerebrospinal fluid (CSF), parenchyma, and the meninges, frequently forming tertiary lymphoid structures in the latter. Several groups, including our own, have shown that B cells from distinct locations within the MS CNS are clonally related and display the characteristics of an antigen-driven response. However, the antigen(s) driving this response have yet to be conclusively defined. To explore the antigen specificity of the MS B cell response, we produced recombinant human immunoglobulin (rIgG) from a series of expanded B cell clones that we isolated from the CNS tissue of six MS brains. The specificity of these MS-derived rIgG and control rIgG derived from non-MS tissues was then examined using multiple methodologies that included testing individual candidate antigens, screening with high-throughput antigen arrays and evaluating binding to CNS-derived cell lines. We report that while several MS-derived rIgG recognized particular antigens, including neurofilament light and a protocadherin isoform, none were unique to MS, as non-MS-derived rIgG used as controls invariably displayed similar binding specificities. We conclude that while MS CNS resident B cells display the characteristics of an antigen-driven B cell response, the antigen(s) driving this response remain at large.
Collapse
Affiliation(s)
- Simon N Willis
- Department of Neurology, Yale School of Medicine , New Haven, CT , USA ; Walter and Eliza Hall Institute of Medical Research , Parkville, VIC , Australia ; Department of Medical Biology, University of Melbourne , Parkville, VIC , Australia
| | | | - Anne Chastre
- Department of Neurology, Yale School of Medicine , New Haven, CT , USA
| | - Shannon D Compton
- Department of Neurology, Yale School of Medicine , New Haven, CT , USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine , New Haven, CT , USA ; Department of Immunobiology, Yale School of Medicine , New Haven, CT , USA
| | - Kevin C O'Connor
- Department of Neurology, Yale School of Medicine , New Haven, CT , USA
| |
Collapse
|
25
|
Stern JNH, Yaari G, Vander Heiden JA, Church G, Donahue WF, Hintzen RQ, Huttner AJ, Laman JD, Nagra RM, Nylander A, Pitt D, Ramanan S, Siddiqui BA, Vigneault F, Kleinstein SH, Hafler DA, O'Connor KC. B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci Transl Med 2015; 6:248ra107. [PMID: 25100741 DOI: 10.1126/scitranslmed.3008879] [Citation(s) in RCA: 327] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) characterized by autoimmune-mediated demyelination and neurodegeneration. The CNS of patients with MS harbors expanded clones of antigen-experienced B cells that reside in distinct compartments including the meninges, cerebrospinal fluid (CSF), and parenchyma. It is not understood whether this immune infiltrate initiates its development in the CNS or in peripheral tissues. B cells in the CSF can exchange with those in peripheral blood, implying that CNS B cells may have access to lymphoid tissue that may be the specific compartment(s) in which CNS-resident B cells encounter antigen and experience affinity maturation. Paired tissues were used to determine whether the B cells that populate the CNS mature in the draining cervical lymph nodes (CLNs). High-throughput sequencing of the antibody repertoire demonstrated that clonally expanded B cells were present in both compartments. Founding members of clones were more often found in the draining CLNs. More mature clonal members derived from these founders were observed in the draining CLNs and also in the CNS, including lesions. These data provide new evidence that B cells traffic freely across the tissue barrier, with the majority of B cell maturation occurring outside of the CNS in the secondary lymphoid tissue. Our study may aid in further defining the mechanisms of immunomodulatory therapies that either deplete circulating B cells or affect the intrathecal B cell compartment by inhibiting lymphocyte transmigration into the CNS.
Collapse
Affiliation(s)
- Joel N H Stern
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Gur Yaari
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA. Bioengineering Program, Faculty of Engineering, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Jason A Vander Heiden
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA
| | - George Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Rogier Q Hintzen
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, and MS Centrum ErasMS, 3000 CA Rotterdam, the Netherlands
| | - Anita J Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Jon D Laman
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, and MS Centrum ErasMS, 3000 CA Rotterdam, the Netherlands
| | - Rashed M Nagra
- Neurology Research, West Los Angeles VA Medical Center, Los Angeles, CA 90073, USA
| | - Alyssa Nylander
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Sriram Ramanan
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Bilal A Siddiqui
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Francois Vigneault
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA. AbVitro Incorporated, Boston, MA 02210, USA
| | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA. Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA.
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA. Department of Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA.
| | - Kevin C O'Connor
- Department of Neurology, Yale School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
26
|
Mastorodemos V, Ioannou M, Verginis P. Cell-based modulation of autoimmune responses in multiple sclerosis and experimental autoimmmune encephalomyelitis: therapeutic implications. Neuroimmunomodulation 2015; 22:181-95. [PMID: 24852748 DOI: 10.1159/000362370] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/20/2014] [Indexed: 11/19/2022] Open
Abstract
Multiple sclerosis (MS) is a prototypic autoimmune inflammatory disorder of the central nervous system (CNS). MS pathogenesis is a complex phenomenon that is influenced by genetic and environmental factors that lead to the dysregulation of immune homeostasis and tolerance. It has been shown that pathogenic T lymphocyte subsets, such as T helper 1 (Th1) and Th17 cells, play a crucial role in the autoimmune cascade influencing disease initiation, progression and subsequent tissue damage during MS. On the other hand, several mechanisms have been described in both patients and animal models of MS with the potential to modulate myelin-specific autoimmune responses and to facilitate amelioration of disease pathology. To this end, regulatory T cells (Tregs) are considered to be a powerful cell subset not only in the maintenance of homeostasis but also in the re-establishment of tolerance. Along these lines, other cell subsets such as dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), γδ T cells and natural killer (NK) cells have been shown to regulate the autoimmune response in the CNS under certain circumstances. This review will attempt to summarize the relevant knowledge of the regulatory mechanisms exerted by immune cells in MS that could hold the promise for the design of novel therapeutic strategies.
Collapse
|
27
|
Abstract
BACKGROUND Many aspects of autoimmune disease are not well understood, including the specificities of autoimmune targets, and patterns of co-morbidity and cross-heritability across diseases. Prior work has provided evidence that somatic mutation caused by gene conversion and deletion at segmentally duplicated loci is relevant to several diseases. Simple tandem repeat (STR) sequence is highly mutable, both somatically and in the germ-line, and somatic STR mutations are observed under inflammation. RESULTS Protein-coding genes spanning STRs having markers of mutability, including germ-line variability, high total length, repeat count and/or repeat similarity, are evaluated in the context of autoimmunity. For the initiation of autoimmune disease, antigens whose autoantibodies are the first observed in a disease, termed primary autoantigens, are informative. Three primary autoantigens, thyroid peroxidase (TPO), phogrin (PTPRN2) and filaggrin (FLG), include STRs that are among the eleven longest STRs spanned by protein-coding genes. This association of primary autoantigens with long STR sequence is highly significant (p<3.0x10(-7)). Long STRs occur within twenty genes that are associated with sixteen common autoimmune diseases and atherosclerosis. The repeat within the TTC34 gene is an outlier in terms of length and a link with systemic lupus erythematosus is proposed. CONCLUSIONS The results support the hypothesis that many autoimmune diseases are triggered by immune responses to proteins whose DNA sequence mutates somatically in a coherent, consistent fashion. Other autoimmune diseases may be caused by coherent somatic mutations in immune cells. The coherent somatic mutation hypothesis has the potential to be a comprehensive explanation for the initiation of many autoimmune diseases.
Collapse
Affiliation(s)
- Kenneth Andrew Ross
- Department of Computer Science, Columbia University, New York, New York, United States of America
| |
Collapse
|
28
|
Cantor JM. CD98 is a potential target for ablating B cell clonal expansion and autoantibody in multiple sclerosis. J Neuroimmunol 2014; 274:230-3. [PMID: 25002078 DOI: 10.1016/j.jneuroim.2014.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/01/2014] [Accepted: 06/17/2014] [Indexed: 01/15/2023]
Abstract
Current B cell-directed therapies for multiple sclerosis impact multiple B cell functions. CD98hc enables B cell clonal expansion and antibody production. I probed the relative importance of autoantibody secretion vs. other B cell functions in MS and targeted CD98hc as a possible therapeutic strategy. I report that the loss of CD98hc function in B cells largely prevents autoantibody production while preserving antigen-presenting and T cell-directing capacities. Mice lacking CD98hc in B cells are protected from EAE; importantly this is overcome with autoantibody-containing plasma. Thus CD98hc blockade is a possible avenue to treat MS by inhibiting clonal expansion and autoantibody.
Collapse
Affiliation(s)
- Joseph M Cantor
- Department of Medicine, University of California San Diego, MC 0726, 9500 Gilman Drive, La Jolla, CA 92093-0726, United States.
| |
Collapse
|
29
|
Schirmer L, Srivastava R, Kalluri SR, Böttinger S, Herwerth M, Carassiti D, Srivastava B, Gempt J, Schlegel J, Kuhlmann T, Korn T, Reynolds R, Hemmer B. Differential loss of KIR4.1 immunoreactivity in multiple sclerosis lesions. Ann Neurol 2014; 75:810-28. [DOI: 10.1002/ana.24168] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 04/26/2014] [Accepted: 04/26/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Lucas Schirmer
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Rajneesh Srivastava
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Sudhakar Reddy Kalluri
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Susanne Böttinger
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Marina Herwerth
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Daniele Carassiti
- Wolfson Neuroscience Laboratories, Division of Brain Sciences, Imperial College Faculty of Medicine, Hammersmith Hospital; London United Kingdom
| | - Barkha Srivastava
- Comprehensive Pneumology Center, Ludwig Maximilians University Munich and Helmholtz Center Munich; Munich Germany
| | - Jens Gempt
- Department of Neurosurgery; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Jürgen Schlegel
- Division of Neuropathology, Institute of Pathology, Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster; Münster Germany
| | - Thomas Korn
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Munich Cluster for Systems Neurology (SyNergy); Munich Germany
| | - Richard Reynolds
- Wolfson Neuroscience Laboratories, Division of Brain Sciences, Imperial College Faculty of Medicine, Hammersmith Hospital; London United Kingdom
| | - Bernhard Hemmer
- Department of Neurology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Munich Cluster for Systems Neurology (SyNergy); Munich Germany
| |
Collapse
|
30
|
Schirmer L, Srivastava R, Hemmer B. To look for a needle in a haystack: the search for autoantibodies in multiple sclerosis. Mult Scler 2014; 20:271-9. [DOI: 10.1177/1352458514522104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The search for autoantibodies in multiple sclerosis (MS) has been challenging for the last 3 decades. With the development of new proteomic methods and advances in expression and assay technologies, progress in the identification of MS autoantibodies has been made. A number of MS-specific autoantibodies have been proposed, most of them targeting proteins expressed in oligodendrocytes and along the myelin sheath. In this review, we summarize the status of antibody research in MS and then discuss recent developments and future strategies in defining and characterizing the potential antigenic targets of autoantibodies in MS.
Collapse
Affiliation(s)
- Lucas Schirmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Rajneesh Srivastava
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany
- Munich Cluster for Systems Neurology (SyNergy), Germany
| |
Collapse
|
31
|
The experimental autoimmune encephalomyelitis (EAE) model of MS: utility for understanding disease pathophysiology and treatment. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:173-89. [PMID: 24507518 DOI: 10.1016/b978-0-444-52001-2.00008-x] [Citation(s) in RCA: 319] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While no single model can exactly recapitulate all aspects of multiple sclerosis (MS), animal models are essential in understanding the induction and pathogenesis of the disease and to develop therapeutic strategies that limit disease progression and eventually lead to effective treatments for the human disease. Several different models of MS exist, but by far the best understood and most commonly used is the rodent model of experimental autoimmune encephalomyelitis (EAE). This model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. Mouse models are most frequently used because of the inbred genotype of laboratory mice, their rapid breeding capacity, the ease of genetic manipulation, and availability of transgenic and knockout mice to facilitate mechanistic studies. Although not all therapeutic strategies for MS have been developed in EAE, all of the current US Food and Drug Administration (FDA)-approved immunomodulatory drugs are effective to some degree in treating EAE, a strong indicator that EAE is an extremely useful model to study potential treatments for MS. Several therapies, such as glatiramer acetate (GA: Copaxone), and natalizumab (Tysabri), were tested first in the mouse model of EAE and then went on to clinical trials. Here we discuss the usefulness of the EAE model in understanding basic disease pathophysiology and developing treatments for MS as well as the potential drawbacks of this model.
Collapse
|
32
|
Anti-brain autoantibodies in the serum of schizophrenic patients: a case-control study. Psychiatry Res 2013; 210:800-5. [PMID: 24103908 DOI: 10.1016/j.psychres.2013.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 06/04/2013] [Accepted: 09/08/2013] [Indexed: 11/23/2022]
Abstract
Schizophrenia is considered a neurodevelopmental disorder with a multifactorial pathogenesis where autoimmune factors may play a significant role. The aim of this study was to verify the presence of anti-brain autoantibodies in the serum of schizophrenic patients compared to healthy controls. Autoantibodies against brain were detected by the immunofluorescence method, utilizing sections of rat hippocampus and hypothalamus and of monkey cerebellum. Three different fluorescence patterns were observed, staining the nucleus-cytoplasm of neurons, the neuroendothelial of blood vessel and the neurofilaments. Search for other organ-specific and non organ-specific autoantibodies was performed in all sera by indirect immunofluorescence method, enzyme linked immunosorbent assay and chemiluminescence immunoassay. Results showed a significant association between schizophrenia and anti-brain autoantibodies against the neuroendothelium of blood vessel in hypothalamus, hippocampus and cerebellum; a significant nuclear and cytoplasmic staining of neurons was assessed only for the hippocampus. No other significant association was found, except between schizophrenia and anti-nuclear autoantibodies on HEp-2 cells. In conclusion, these results support the hypothesis of a significant association between schizophrenia and circulating anti-brain autoantibodies, suggesting a diffuse reactivity against the neuroendothelium of blood vessel and highlighting a nuclear and cytoplasmic staining of the neurons of hippocampus.
Collapse
|
33
|
Wang KC, Lee CL, Chen SY, Chen JC, Yang CW, Chen SJ, Tsai CP. Distinct serum cytokine profiles in neuromyelitis optica and multiple sclerosis. J Interferon Cytokine Res 2013; 33:58-64. [PMID: 23398365 DOI: 10.1089/jir.2012.0040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common prototypic inflammatory demyelinating disease. Neuromyelitis optica (NMO) is another inflammatory demyelinating disease of the central nervous system that exhibits clinical symptoms mainly associated with optic neuritis and myelopathy. The inflammatory reaction in MS is associated with an upregulation of a variety of T helper 1 (Th1)- or Th17-mediated cytokines. However, NMO and MS are intertwined both clinically and pathologically, which complicates their diagnosis and treatment. The aim of this study was to evaluate the differences in serum cytokine levels in patients with NMO and MS. We collected peripheral serum from patients with these central nervous system demyelinating diseases for the study. A cytometric bead array was used to assess the cytokine levels using flow cytometry. We found more inflammatory [interleukin (IL)-2 and interferon-γ) and anti-inflammatory (IL-4 and IL-10) cytokines in NMO than in MS. The differences in the optimal cutoff points of serum cytokines, including IL-2 ≥5 pg/mL, can differentiate NMO from MS. In conclusion, patients with NMO had an increased Th1-mediated inflammatory response, but similar Th17-mediated inflammation changes compared to patients with MS. Serum cytokine studies can differentiate NMO cases from MS.
Collapse
Affiliation(s)
- Kai Chen Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
34
|
Querol L, Clark PL, Bailey MA, Cotsapas C, Cross AH, Hafler DA, Kleinstein SH, Lee JY, Yaari G, Willis SN, O'Connor KC. Protein array-based profiling of CSF identifies RBPJ as an autoantigen in multiple sclerosis. Neurology 2013; 81:956-63. [PMID: 23921886 DOI: 10.1212/wnl.0b013e3182a43b48] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To profile the reactivity of CSF-derived immunoglobulin from patients with multiple sclerosis (MS) against a large panel of antigens, to identify disease-specific reactivities. METHODS CSF from subjects with MS with elevated immunoglobulin G and CSF from control subjects presenting with other inflammatory neurologic disease were screened against a protein array consisting of 9,393 proteins. Reactivity to a candidate protein identified using these arrays was confirmed with ELISA and immunocytochemistry. RESULTS Autoantibodies against one protein on the array, recombination signal binding protein for immunoglobulin kappa J region (RBPJ), discriminated between patients with MS and controls (p = 0.0052). Using a large validation cohort, we found a higher prevalence of autoantibodies against RBPJ in the CSF of patients with MS (12.5%) compared with the CSF of patients with other neurologic diseases (1.6%; p = 0.02) by ELISA. This difference in reactivity was restricted to the CSF as serum reactivity against RBPJ did not differ between patients and controls. The presence of CSF autoantibodies against RBPJ was further confirmed by immunocytochemistry. CONCLUSIONS These data indicate that RBPJ, a ubiquitous protein of the Notch signaling pathway that plays an important role in Epstein-Barr virus infection, is a novel MS autoantigen candidate that is recognized by CSF-derived immunoglobulin G in a subset of patients with MS.
Collapse
Affiliation(s)
- Luis Querol
- From the Department of Neurology (L.Q., P.L.C., M.A.B., C.C., D.A.H., J.-Y.L., K.C.O.), Human and Translational Immunology Program (D.A.H., K.C.O.), Department of Genetics (C.C.), Department of Pathology (S.H.K., G.Y.), and Department of Immunobiology (D.A.H.), Yale School of Medicine, New Haven, CT; Neuromuscular Diseases Unit (L.Q.), Hospital de la Santa Creu i Sant Pau, Universitat Autónoma de Barcelona, Spain; Medical and Population Genetics (C.C.), Broad Institute of MIT and Harvard, Cambridge, MA; Department of Neurology (A.H.C.), Washington University School of Medicine, St. Louis, MO; Interdepartmental Program in Computational Biology and Bioinformatics (S.H.K.), Yale University, New Haven, CT; and Department of Neurology (S.N.W.), Harvard Medical School and Brigham and Women's Hospital, Boston, MA. Simon N. Willis is currently affiliated with the Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Cappellano G, Carecchio M, Fleetwood T, Magistrelli L, Cantello R, Dianzani U, Comi C. Immunity and inflammation in neurodegenerative diseases. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:89-107. [PMID: 23844334 PMCID: PMC3703122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 06/11/2013] [Indexed: 06/02/2023]
Abstract
Immune reactions inside the central nervous system are finely regulated, thanks to the presence of several checkpoints that have the fundamental purpose to preserve this fragile tissue form harmful events. The current knowledge on the role of neuroinflammation and neuro-immune interactions in the fields of multiple sclerosis, Alzheimer's disease and Parkinson's disease is reviewed. Moreover, a focus on the potential role of both active and passive immunotherapy is provided. Finally, we propose a common perspective, which implies that, under pathological conditions, inflammation may exert both detrimental and protective functions, depending on local factors and the timing of immune activation and shutting-off systems.
Collapse
Affiliation(s)
- Giuseppe Cappellano
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Health Sciences, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Miryam Carecchio
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Thomas Fleetwood
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Luca Magistrelli
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Roberto Cantello
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Health Sciences, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| | - Cristoforo Comi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
- Department of Translational Medicine, Section of Neurology, University of Eastern Piedmont, “Amedeo Avogadro”Novara, Italy
| |
Collapse
|
36
|
Pedotti R, Musio S, Scabeni S, Farina C, Poliani PL, Colombo E, Costanza M, Berzi A, Castellucci F, Ciusani E, Confalonieri P, Hemmer B, Mantegazza R, Antozzi C. Exacerbation of experimental autoimmune encephalomyelitis by passive transfer of IgG antibodies from a multiple sclerosis patient responsive to immunoadsorption. J Neuroimmunol 2013; 262:19-26. [PMID: 23768729 DOI: 10.1016/j.jneuroim.2013.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/10/2013] [Accepted: 05/17/2013] [Indexed: 10/26/2022]
Abstract
The pathogenic role of antibodies in multiple sclerosis (MS) is still controversial. We transferred to mice with experimental autoimmune encephalomyelitis (EAE), animal model of MS, IgG antibodies purified from a MS patient presenting a dramatic clinical improvement during relapse after selective IgG removal with immunoadsorption. Passive transfer of patient's IgG exacerbated motor paralysis and increased mouse central nervous system (CNS) inflammation and demyelination. Binding of patient's IgG was demonstrated in mouse CNS, with a diffuse staining of white matter oligodendrocytes. These data support a growing body of evidence that antibodies can play an important role in the pathobiology of MS.
Collapse
Affiliation(s)
- Rosetta Pedotti
- Neuroimmunology and Neuromuscular Disorders Unit, Foundation IRCCS Neurological Institute C. Besta, Via Celoria 11, 20133 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Tolerogenic vaccines represent a new class of vaccine designed to re-establish immunological tolerance, restore immune homeostasis, and thereby reverse autoimmune disease. Tolerogenic vaccines induce long-term, antigen-specific, inhibitory memory that blocks pathogenic T cell responses via loss of effector T cells and gain of regulatory T cell function. Substantial advances have been realized in the generation of tolerogenic vaccines that inhibit experimental autoimmune encephalomyelitis in a preclinical setting, and these vaccines may be a prequel of the tolerogenic vaccines that may have therapeutic benefit in Multiple Sclerosis. The purpose here is to provide a snapshot of the current concepts and future prospects of tolerogenic vaccination for Multiple Sclerosis, along with the central challenges to clinical application.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | | |
Collapse
|
38
|
Toubi E, Nussbaum S, Staun-Ram E, Snir A, Melamed D, Hayardeny L, Miller A. Laquinimod modulates B cells and their regulatory effects on T cells in multiple sclerosis. J Neuroimmunol 2012; 251:45-54. [PMID: 22846497 DOI: 10.1016/j.jneuroim.2012.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 11/19/2022]
Abstract
Laquinimod is an orally administered drug under development for the treatment of Multiple Sclerosis (MS), lacking a fully elucidated mode of action. We assessed the immunomodulatory effects of laquinimod in vitro on human B cells from healthy or MS patients, cultured alone or with CD4(+) T cells. Laquinimod modulated B cell markers, mainly by increasing the regulatory ones CD25, IL10 and CD86, and decreased IL4, while increasing IL10 and TGFβ in both B and T cells, in a B cell-mediated manner. These findings shed additional light on the mechanisms underlying the effects of laquinimod in MS and potentially other immune-mediated diseases.
Collapse
Affiliation(s)
- Elias Toubi
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
39
|
Geherin SA, Fintushel SR, Lee MH, Wilson RP, Patel RT, Alt C, Young AJ, Hay JB, Debes GF. The skin, a novel niche for recirculating B cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:6027-35. [PMID: 22561151 DOI: 10.4049/jimmunol.1102639] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
B cells infiltrate the skin in many chronic inflammatory diseases caused by autoimmunity or infection. Despite potential contribution to disease, skin-associated B cells remain poorly characterized. Using an ovine model of granulomatous skin inflammation, we demonstrate that B cells increase in the skin and skin-draining afferent lymph during inflammation. Surprisingly, skin B cells are a heterogeneous population that is distinct from lymph node B cells, with more large lymphocytes as well as B-1-like B cells that coexpress high levels of IgM and CD11b. Skin B cells have increased MHC class II, CD1, and CD80/86 expression compared with lymph node B cells, suggesting that they are well-suited for T cell activation at the site of inflammation. Furthermore, we show that skin accumulation of B cells and Ab-secreting cells during inflammation increases local Ab titers, which could augment host defense and autoimmunity. Although skin B cells express typical skin-homing receptors, such as E-selectin ligand and α-4 and β-1 integrins, they are unresponsive to ligands for chemokine receptors associated with T cell homing into skin. Instead, skin B cells migrate toward the cutaneously expressed CCR6 ligand CCL20. Our data support a model in which B cells use CCR6-CCL20 to recirculate through the skin, fulfilling a novel role in skin immunity and inflammation.
Collapse
Affiliation(s)
- Skye A Geherin
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
The advent of monoclonal antibodies in the treatment of chronic autoimmune diseases. Neurol Sci 2011; 31 Suppl 3:283-8. [PMID: 20644974 DOI: 10.1007/s10072-010-0382-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Monoclonal antibodies, first introduced in cancer therapy and to prevent allograft rejection, represent new pharmacological tools for the treatment of autoimmune diseases. With the knowledge of immunological movements in autoimmunity, it is now possible to target each single step of the immune process, from the activation of T lymphocytes in lymph nodes to the formation of the immunological synapse, and to T cell differentiation and cytokine production. However, this approach is still not devoid of adverse effects. In fact, even if monoclonal antibodies exert selective immunomodulation by targeting only cells expressing a specific antigen, a widespread perturbation of the immune system is induced, leading to a predisposition for infections and infestations and to the occurrence of tumours.
Collapse
|
41
|
Kiraly A, Koffman B, Hacker M, Gunning W, Rasche S, Quinn A. A novel aza-anthrapyrazole blocks the progression of experimental autoimmune encephalomyelitis after the priming of autoimmunity. Clin Immunol 2011; 141:304-16. [DOI: 10.1016/j.clim.2011.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/17/2011] [Accepted: 08/18/2011] [Indexed: 11/16/2022]
|
42
|
Targeting poly(ADP-ribose) polymerase-1 as a promising approach for immunomodulation in multiple sclerosis? Trends Mol Med 2011; 18:92-100. [PMID: 22078487 DOI: 10.1016/j.molmed.2011.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/27/2011] [Accepted: 10/06/2011] [Indexed: 12/23/2022]
Abstract
Despite significant advancement in developing therapies for multiple sclerosis (MS), drugs that cure this devastating disorder are an unmet need. Among the remedies showing efficacy in preclinical MS models, inhibitors of poly(ADP-ribose) polymerase (PARP)-1 have gained great momentum. Emerging evidence demonstrates that PARP-1 inhibitors epigenetically regulate gene expression and finely tune transcriptional activation in immune and neural cells. In this review, we present an appraisal of the effects of PARP-1 and its inhibitors on immune activation, with particular emphasis on the processes taking place during the autoimmune attack directed against the central nervous system. One explanation is that drugs inhibiting PARP-1 activity protect from neuroinflammation in MS models via immunomodulation and direct neuroprotection. PARP-1 inhibitors have already reached the clinical arena as cancer treatments, and observations made in treating these patients could help advance treatments for MS.
Collapse
|
43
|
Kapadia M, Sakic B. Autoimmune and inflammatory mechanisms of CNS damage. Prog Neurobiol 2011; 95:301-33. [DOI: 10.1016/j.pneurobio.2011.08.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/13/2022]
|
44
|
Kosmidis ML, Dalakas MC. Practical considerations on the use of rituximab in autoimmune neurological disorders. Ther Adv Neurol Disord 2011; 3:93-105. [PMID: 21179602 DOI: 10.1177/1756285609356135] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rituximab (Mabthera, Rituxan) is a chimeric human/murine monoclonal antibody against CD-20 surface antigen expressed on B-cells. Rituximab, by causing B-cell depletion, appears to be effective in several autoimmune disorders; it has been approved for rheumatoid arthritis and is a promising new agent in the treatment of several autoimmune neurological disorders. A controlled study in patients with relapsing remitting multiple sclerosis has shown that rituximab significantly reduces the number of new MRI lesions and improves clinical outcome; it also showed some promise in a subset of patients with primary progressive MS. The drug is also effective in a number of patients with Devic's disease, myasthenia gravis, autoimmune neuropathies, and inflammatory myopathies. The apparent effectiveness of rituximab has moved B-cells into the center stage of clinical and laboratory investigation of autoimmune neurological disorders. We review the evidence-based effectiveness of rituximab in neurological disorders based on controlled trials and anecdotal reports, including our own experience, and address the immunobiology of B-cells in autoimmune central nervous system (CNS) and peripheral nervous system (PNS) disorders. In addition, we provide practical guidelines on how best to use this drug in clinical practice and highlight its potential toxicity.
Collapse
Affiliation(s)
- Mixalis L Kosmidis
- Neuroimmunology Section, Department of Pathophysiology, University of Athens Medical School, Athens, Greece
| | | |
Collapse
|
45
|
De Jager PL. Identifying patient subtypes in multiple sclerosis and tailoring immunotherapy: challenges for the future. Ther Adv Neurol Disord 2011; 2:8-19. [PMID: 21180626 DOI: 10.1177/1756285609337976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The accelerating pace of technological and analytical development in the fields of genetic and phenotypic profiling has ushered in an era of great promise for multiple sclerosis (MS) research. As we continue to identify modest but meaningful associations to MS susceptibility, disease course, treatment response, and other clinical or paraclinical phenotypes, we must begin to (1) embark on the challenging set of studies that will integrate disparate observations into clinical algorithms, and (2) validate their clinical utility. Genetic data are receiving muchofthe attention today, but they are unlikelytobesufficienttooffer a personalized approach to disease management in MS. Rather, the genetic architecture of the disease, once uncovered, will offer a fixed platform upon which more dynamic molecular profiles can be assembled to deconstruct the structure of the patient population that we label with a diagnosis of MS. The tools and methods to gain insight into the heterogeneity of MS patients are available today; we must now realize their potential in enhancing the care of MS patients.
Collapse
|
46
|
Myelin-reactive antibodies mediate the pathology of MBP–PLP fusion protein MP4-induced EAE. Clin Immunol 2011; 140:54-62. [DOI: 10.1016/j.clim.2011.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 02/21/2011] [Accepted: 03/15/2011] [Indexed: 11/18/2022]
|
47
|
Bartok B, Silverman GJ. Development of anti-CD20 therapy for multiple sclerosis. Exp Cell Res 2011; 317:1312-8. [PMID: 21510932 PMCID: PMC3266104 DOI: 10.1016/j.yexcr.2011.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Revised: 04/01/2011] [Accepted: 04/04/2011] [Indexed: 11/29/2022]
Abstract
The therapeutic utility of the targeting of B lymphocytes is currently being evaluated in a range of autoimmune diseases that include multiple sclerosis (MS). For MS, even though intrathecal immunoglobulin production is a hallmark of multiple sclerosis (MS), T cells have long been considered as the main effectors of pathogenesis. Recognition of the roles of autoreactive B cells has changed this conventional view of the disease and also provided a rationale for studies of anti-CD20 therapy in MS. Recent trials suggest that this approach may provide clinical benefits in some MS patients that equal or surpass currently approved approaches, yet not all patients may benefit. In this review we provide an overview on recent progress on these trials.
Collapse
Affiliation(s)
- Beatrix Bartok
- Department of Medicine, UC San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | |
Collapse
|
48
|
Fontoura P. Monoclonal antibody therapy in multiple sclerosis: Paradigm shifts and emerging challenges. MAbs 2011; 2:670-81. [PMID: 21124072 DOI: 10.4161/mabs.2.6.13270] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Therapeutic approaches to multiple sclerosis (MS) are based on altering the functions of the immune system, either by using broad immunosuppressive drugs used for transplantation rejection and rheumatology, or by modulating them more discreetly with beta interferon and synthetic amino-acid copolymers. These strategies are only partially successful, have important safety and tolerability limitations, and have shown to be mostly effective in earlier stages of the disease, in which acute relapses dominate the clinical picture. For progressive phenotypes of MS there are currently no effective therapeutic options. As very specific and potent immunosuppressive agents, monoclonal antibodies (mAbs) may offer considerable advantages over other therapies for MS. During the last decade, anti-a4 integrin natalizumab became the first approved mAb for treatment of relapsing MS, after convincingly demonstrating clinically significant effects on two large Phase 3 trials. Moreover, the concept of disease remission was introduced for the first time, to describe patients that show no signs of clinical or imaging markers of disease activity during therapy with natalizumab. Of the mAbs under development for MS, alemtuzumab and rituximab have also shown promising evidence of effectiveness, and potentially expanded the therapeutic horizon to reversal of disease progression in early relapsing patients, and progressive patients who previously had not been studied. However, the appearance of progressive multifocal leukoencephalopathy (PML) in natalizumab-treated MS patients, as well as in patients with lymphoma, lupus and rheumatoid arthritis treated with rituximab, and autoimmune-type complications in alemtuzumab-treated MS patients underlines the fact that extended efficacy comes with significant clinical risks. The challenge is then how best to utilize therapies that have evidently superior efficacy in a chronic disease of young adults, to obtain the best benefit-risk ratio, and how to monitor and prevent emergent safety concerns.
Collapse
Affiliation(s)
- Paulo Fontoura
- Roche Pharmaceuticals, Pharma Research and Exploratory Development, Translational Medicine CNS, Basel, Switzerland.
| |
Collapse
|
49
|
Ragheb S, Li Y, Simon K, VanHaerents S, Galimberti D, De Riz M, Fenoglio C, Scarpini E, Lisak R. Multiple sclerosis: BAFF and CXCL13 in cerebrospinal fluid. Mult Scler 2011; 17:819-29. [PMID: 21372118 DOI: 10.1177/1352458511398887] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is increasing evidence of B-cell involvement in the pathogenesis of multiple sclerosis (MS). B-cell activating factor (BAFF) has an essential role in B-cell homeostasis. The chemokine CXCL13 has an important role in the formation and maintenance of B-cell follicles. OBJECTIVE To measure BAFF and CXCL13 levels in the cerebrospinal fluid (CSF) of patients with MS compared to patients with other neurological diseases. METHODS Cytokine/chemokine levels were measured by an enzyme-linked immunosorbent assay (ELISA). RESULTS In MS patients, BAFF levels were highest in patients with secondary progressive disease, and were higher during relapse in patients with relapsing-remitting and secondary progressive disease. CXCL13 levels were also higher during relapse. There was a positive correlation between CXCL13 and the IgG index, and an inverse correlation between BAFF and the IgG index. The implications of this finding are discussed. CONCLUSION During relapse, we found various positive correlations between BAFF, CXCL13 and the cytokines IL-6 and IL-10. These findings show that molecules that are essential for B-cell recruitment, survival, maturation and function may be working in concert to affect B-cell homeostasis in MS and contribute to the pathophysiology of the disease.
Collapse
Affiliation(s)
- Samia Ragheb
- Department of Neurology, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Di Pauli F, Mader S, Rostasy K, Schanda K, Bajer-Kornek B, Ehling R, Deisenhammer F, Reindl M, Berger T. Temporal dynamics of anti-MOG antibodies in CNS demyelinating diseases. Clin Immunol 2011; 138:247-54. [DOI: 10.1016/j.clim.2010.11.013] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 11/18/2010] [Indexed: 01/21/2023]
|