1
|
Behrens M, Comabella M, Lünemann JD. EBV-specific T-cell immunity: relevance for multiple sclerosis. Front Immunol 2024; 15:1509927. [PMID: 39776919 PMCID: PMC11703957 DOI: 10.3389/fimmu.2024.1509927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Genetic and environmental factors jointly determine the susceptibility to develop multiple sclerosis (MS). Improvements in the design of epidemiological studies have helped to identify consistent environmental risk associations such as the increased susceptibility for MS following Epstein-Barr virus (EBV) infection, while biological mechanisms that drive the association between EBV and MS remain incompletely understood. An increased and broadened repertoire of antibody and T-cell immune responses to EBV-encoded antigens, especially to the dominant CD4+ T-cell EBV nuclear antigen 1 (EBNA1), is consistently observed in patients with MS, indicating that protective EBV-specific immune responses are deregulated in MS and potentially contribute to disease development. Exploitation of B-cell trajectories by EBV infection might promote survival of autoreactive B-cell species and proinflammatory B:T-cell interactions. In this review article, we illustrate evidence for a causal role of EBV infection in MS, discuss how EBV-targeting adaptive immune responses potentially modulate disease susceptibility and progression, and provide future perspectives on how novel model systems could be utilized to better define the role of EBV and viral pathogens in MS. Insights gained from these studies might facilitate the development of prevention strategies and more effective treatments for MS.
Collapse
Affiliation(s)
- Malina Behrens
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| |
Collapse
|
2
|
Pressley KR, Schwegman L, De Oca Arena MM, Huizar CC, Zamvil SS, Forsthuber TG. HLA-transgenic mouse models to study autoimmune central nervous system diseases. Autoimmunity 2024; 57:2387414. [PMID: 39167553 PMCID: PMC11470778 DOI: 10.1080/08916934.2024.2387414] [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: 03/31/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/23/2024]
Abstract
It is known that certain human leukocyte antigen (HLA) genes are associated with autoimmune central nervous system (CNS) diseases, such as multiple sclerosis (MS), but their exact role in disease susceptibility and etiopathogenesis remains unclear. The best studied HLA-associated autoimmune CNS disease is MS, and thus will be the primary focus of this review. Other HLA-associated autoimmune CNS diseases, such as autoimmune encephalitis and neuromyelitis optica will be discussed. The lack of animal models to accurately capture the complex human autoimmune response remains a major challenge. HLA transgenic (tg) mice provide researchers with powerful tools to investigate the underlying mechanisms promoting susceptibility and progression of HLA-associated autoimmune CNS diseases, as well as for elucidating the myelin epitopes potentially targeted by T cells in autoimmune disease patients. We will discuss the potential role(s) of autoimmune disease-associated HLA alleles in autoimmune CNS diseases and highlight information provided by studies using HLA tg mice to investigate the underlying pathological mechanisms and opportunities to use these models for development of novel therapies.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | | | - Carol Chase Huizar
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Scott S. Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
3
|
Caragea AM, Ursu RI, Maruntelu I, Tizu M, Constantinescu AE, Tălăngescu A, Constantinescu I. High Resolution HLA-A, HLA-B, and HLA-C Allele Frequencies in Romanian Hematopoietic Stem Cell Donors. Int J Mol Sci 2024; 25:8837. [PMID: 39201523 PMCID: PMC11354460 DOI: 10.3390/ijms25168837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The HLA genes are associated with various autoimmune pathologies, with the control of the immune response also being significant in organs and cells transplantation. The aim of the study is to identify the HLA-A, HLA-B, and HLA-C alleles frequencies in the analyzed Romanian cohort. We performed HLA typing using next-generation sequencing (NGS) in a Romanian cohort to estimate class I HLA allele frequencies up to a six-digit resolution. A total of 420 voluntary donors from the National Registry of Voluntary Hematopoietic Stem Cell Donors (RNDVCSH) were included in the study for HLA genotyping. Peripheral blood samples were taken and brought to the Fundeni Clinical Institute during 2020-2021. HLA genotyping was performed using the Immucor Mia Fora NGS MFlex kit. A total of 109 different alleles were detected in 420 analyzed samples, out of which 31 were for HLA-A, 49 for HLA-B, and 29 for HLA-C. The most frequent HLA-A alleles were HLA-A*02:01:01 (26.11%), HLA-A*01:01:01 (12.5%), HLA-A*24:02:01 (11.67%), HLA-A*03:01:01 (9.72%), HLA-A*11:01:01, and HLA-A*32:01:01 (each with 8.6%). For the HLA-B locus, the most frequent allele was HLA-B*18:01:01 (11.25%), followed by HLA-B*51:01:01 (10.83%) and HLA-B*08:01:01 (7.78%). The most common HLA-C alleles were HLA-C*07:01:01 (17.36%), HLA-C*04:01:01 (13.47%), and HLA-C*12:03:01 (10.69%). Follow-up studies are ongoing for confirming the detected results.
Collapse
Affiliation(s)
- Andreea Mirela Caragea
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
- Center for Immunogenetics and Virology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Radu-Ioan Ursu
- Department of Medical Genetics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ion Maruntelu
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
- Center for Immunogenetics and Virology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Maria Tizu
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
- Center for Immunogenetics and Virology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Alexandra-Elena Constantinescu
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
| | - Adriana Tălăngescu
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
- Center for Immunogenetics and Virology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Ileana Constantinescu
- Department of Immunology and Transplantation Immunology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania; (A.M.C.); (I.M.); (M.T.); (A.-E.C.); (A.T.); (I.C.)
- Center for Immunogenetics and Virology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
4
|
Qi C, Ren H, Fan Y. Microglia specific alternative splicing alterations in multiple sclerosis. Aging (Albany NY) 2024; 16:11656-11667. [PMID: 39115871 PMCID: PMC11346782 DOI: 10.18632/aging.206045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Several aberrant alternative splicing (AS) events and their regulatory mechanisms are widely recognized in multiple sclerosis (MS). Yet the cell-type specific AS events have not been extensively examined. Here we assessed the diversity of AS events using web-based RNA-seq data of sorted CD15-CD11b+ microglia in white matter (WM) region from 10 patients with MS and 11 control subjects. The GSE111972 dataset was downloaded from GEO and ENA databases, aligned to the GRCh38 reference genome from ENSEMBL via STAR. rMATS was used to assess five types of AS events, alternative 3'SS (A3SS), alternative 5'SS (A5SS), skipped exon (SE), retained intron (RI) and mutually exclusive exons (MXE), followed by visualizing with rmats2sashimiplot and maser. Differential genes or transcripts were analyzed using the limma R package. Gene ontology (GO) analysis was performed with the clusterProfiler R package. 42,663 raw counts of AS events were identified and 132 significant AS events were retained based on the filtered criteria: 1) average coverage >10 and 2) delta percent spliced in (ΔPSI) >0.1. SE was the most common AS event (36.36%), followed by MXE events (32.58%), and RI (18.94%). Genes related to telomere maintenance and organization primarily underwent SE splicing, while genes associated with protein folding and mitochondrion organization were predominantly spliced in the MXE pattern. Conversely, genes experiencing RI were enriched in immune response and immunoglobulin production. In conclusion, we identified microglia-specific AS changes in the white matter of MS patients, which may shed light on novel pathological mechanisms underlying MS.
Collapse
Affiliation(s)
- Caiyun Qi
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Keehn CC, Yazdian A, Hunt PJ, Davila-Siliezar P, Laylani NA, Lee AG. Monoclonal antibodies in neuro-ophthalmology. Saudi J Ophthalmol 2024; 38:13-24. [PMID: 38628411 PMCID: PMC11017005 DOI: 10.4103/sjopt.sjopt_256_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 04/19/2024] Open
Abstract
Neuro-ophthalmologic diseases include a broad range of disorders affecting the afferent and efferent visual pathways. Recently, monoclonal antibody (mAb) therapies have emerged as a promising targeted approach in the management of several of these complex conditions. Here, we describe the mechanism-specific applications and advancements in neuro-ophthalmologic mAb therapies. The application of mAbs in neuro-ophthalmologic diseases highlights our increasing understanding of disease-specific mechanisms in autoimmune conditions such as neuromyelitis optica, thyroid eye disease, and myasthenia gravis. Due to the specificity of mAb therapies, applications in neuro-ophthalmologic diseases have yielded exceptional clinical outcomes, including both reduced rate of relapse and progression to disability, visual function preservation, and quality of life improvement. These advancements have not only expanded the range of treatable neuro-ophthalmologic diseases but also reduced adverse events and increased the response rate to treatment. Further research into neuro-ophthalmologic disease mechanisms will provide accurate and specific targeting of important disease mediators through applications of future mAbs. As our understanding of these diseases and the relevant therapeutic targets evolve, we will continue to build on our understanding of how mAbs interfere with disease pathogenesis, and how these changes improve clinical outcomes and quality of life for patients.
Collapse
Affiliation(s)
- Caroline C. Keehn
- Department of Ophthalmology, Baylor College of Medicine, Houston, USA
| | - Arman Yazdian
- Department of Ophthalmology, Baylor College of Medicine, Houston, USA
| | - Patrick J. Hunt
- Department of Ophthalmology, Baylor College of Medicine, Houston, USA
| | - Pamela Davila-Siliezar
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, USA
| | - Noor A. Laylani
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, USA
| | - Andrew G. Lee
- Department of Ophthalmology, Baylor College of Medicine, Houston, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, USA
- Department of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, USA
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, USA
- Department of Ophthalmology, Texas A and M College of Medicine, Bryan, Texas, USA
- Department of Ophthalmology, University of Buffalo, Buffalo, NY, USA
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
6
|
James LM, Georgopoulos AP. Positive Association Between the Immunogenetic Human Leukocyte Antigen (HLA) Profiles of Multiple Sclerosis and Brain Cancer. Neurosci Insights 2023; 18:26331055231214543. [PMID: 38046672 PMCID: PMC10693228 DOI: 10.1177/26331055231214543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Previous research has documented elevated risk of brain cancer in patients with multiple sclerosis (MS). Separately, human leukocyte antigen (HLA) has been implicated in protection or susceptibility for both conditions. The aim of the current study was to assess a possible role of shared immunogenetic influence on risk of MS and brain cancer. We first identified an immunogenetic profile for each condition based on the covariance between the population frequency of 127 high-resolution HLA alleles and the population prevalence of each condition in 14 Continental Western European countries and then evaluated the correspondence between MS and brain cancer immunogenetic profiles. Also, since each individual carries 12 HLA alleles (2 × 6 genes), we estimated HLA protection and susceptibility for MS and brain cancer at the individual level. We found that the immunogenetic profiles of MS and brain cancer were highly correlated overall (P < .001) and across all 6 HLA genes with the strongest association observed for DRB1, followed by DQB1 and HLA-A. These findings of immunogenetic overlap between MS and brain cancer are discussed in light of the role of HLA in the immune system response to viruses and other foreign antigens.
Collapse
Affiliation(s)
- Lisa M James
- Department of Veterans Affairs Health Care System, The HLA Research Group, Brain Sciences Center, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Apostolos P Georgopoulos
- Department of Veterans Affairs Health Care System, The HLA Research Group, Brain Sciences Center, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
7
|
Liu PJ, Yang TT, Fan ZX, Yuan GB, Ma L, Wang ZY, Lu JF, Yuan BY, Zou WL, Zhang XH, Liu GZ. Characterization of antigen-specific CD8+ memory T cell subsets in peripheral blood of patients with multiple sclerosis. Front Immunol 2023; 14:1110672. [PMID: 37215118 PMCID: PMC10192904 DOI: 10.3389/fimmu.2023.1110672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Background Increasing evidence indicates the importance of CD8+ T cells in autoimmune attack against CNS myelin and axon in multiple sclerosis (MS). Previous research has also discovered that myelin-reactive T cells have memory phenotype functions in MS patients. However, limited evidence is available regarding the role of CD8+ memory T cell subsets in MS. This study aimed to explore potential antigen-specific memory T cell-related biomarkers and their association with disease activity. Methods The myelin oligodendrocyte glycoprotein (MOG)-specific CD8+ memory T cell subsets and their related cytokines (perforin, granzyme B, interferon (IFN)-γ) and negative co-stimulatory molecules (programmed cell death protein 1 (PD-1), T- cell Ig and mucin domain 3 (Tim-3)) were analyzed by flow cytometry and real-time PCR in peripheral blood of patients with relapsing-remitting MS. Results We found that MS patients had elevated frequency of MOG-specific CD8+ T cells, MOG-specific central memory T cells (TCM), MOG-specific CD8+ effector memory T cells (TEM), and MOG-specific CD8+ terminally differentiated cells (TEMRA); elevated granzyme B expression on MOG-specific CD8+ TCM; and, on MOG-specific CD8+ TEM, elevated granzyme B and reduced PD-1 expression. The Expanded Disability Status Scale score (EDSS) in MS patients was correlated with the frequency of MOG-specific CD8+ TCM, granzyme B expression in CD8+ TCM, and granzyme B and perforin expression on CD8+ TEM, but with reduced PD-1 expression on CD8+ TEM. Conclusion The dysregulation of antigen-specific CD8+ memory T cell subsets, along with the abnormal expression of their related cytokines and negative co-stimulatory molecules, may reflect an excessive or persistent inflammatory response induced during early stages of the illness. Our findings strongly suggest positive regulatory roles for memory T cell populations in MS pathogenesis, probably via molecular mimicry to trigger or promote abnormal peripheral immune responses. Furthermore, downregulated PD-1 expression may stimulate a positive feedback effect, promoting MS-related inflammatory responses via the interaction of PD-1 ligands. Therefore, these parameters are potential serological biomarkers for predicting disease development in MS.
Collapse
Affiliation(s)
- Pen-Ju Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ze-Xin Fan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Bin Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lin Ma
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ze-Yi Wang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jian-Feng Lu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bo-Yi Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wen-Long Zou
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xing-Hu Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Plafker SM, Titcomb T, Zyla-Jackson K, Kolakowska A, Wahls T. Overview of diet and autoimmune demyelinating optic neuritis: a narrative review. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00022. [PMID: 37128292 PMCID: PMC10144304 DOI: 10.1097/in9.0000000000000022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/29/2023] [Indexed: 05/03/2023]
Abstract
This review summarizes the cellular and molecular underpinnings of autoimmune demyelinating optic neuritis (ADON), a common sequela of multiple sclerosis and other demyelinating diseases. We further present nutritional interventions tested for people with multiple sclerosis focusing on strategies that have shown efficacy or associations with disease course and clinical outcomes. We then close by discuss the potential dietary guidance for preventing and/or ameliorating ADON.
Collapse
Affiliation(s)
- Scott M. Plafker
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tyler Titcomb
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Katarzyna Zyla-Jackson
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Aneta Kolakowska
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Terry Wahls
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
9
|
Mehta RS, Cao K, Saliba RM, Al-Atrash G, Alousi AM, Lontos K, Marcoux C, Carmazzi Y, Rondon G, Bashir Q, Hosing CM, Kebriaei P, Khouri I, Marin D, Nieto Y, Oran B, Popat UR, Qazilbash MH, Ramdial J, Rezvani K, Champlin RE, Shpall EJ. HLA Factors versus Non-HLA Factors for Haploidentical Donor Selection. Transplant Cell Ther 2023; 29:189-198. [PMID: 36470579 PMCID: PMC10125001 DOI: 10.1016/j.jtct.2022.11.027] [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: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
When multiple haploidentical donors are available for transplantation, those of younger generations are generally selected over those of older generations. However, it is unclear who is the optimal donor when selecting candidates from within a generation, such as father versus mother, son versus daughter, or brother versus sister. Although traditionally male donors are favored over female donors, particularly for male recipients, and significant associations of individual HLA mis(matches) on outcomes are being increasingly recognized, the hierarchy of factors for donor selection is indeterminate. To assess whether HLA factors take precedence over non-HLA factors and to isolate the influence of specific characteristics on outcomes, we analyzed 412 patients stratified by donor relationship: child donor (son [n = 202] versus daughter [n = 96]), parent (father [n = 28] versus mother [n = 29]), and sibling (noninherited maternal [NIMA; n = 29] versus paternal [NIPA; n = 28] mismatched). Among siblings, NIMA mismatch was associated with a lower risk of acute graft-versus-host disease (aGVHD); B-leader mismatch was associated with high nonrelapse mortality (NRM), poor progression-free survival, and a trend toward poor overall survival (OS), whereas A-mismatch was associated with lower aGVHD. Among parent donors, the relationship did not impact any outcome; B-leader mismatch was associated with higher NRM and a trend toward poor OS, whereas A-mismatch was associated with lower NRM and improved progression-free survival and OS. Among child donors, no individual HLA mismatch was predictive of any outcome, and daughter donors were not associated with any adverse outcomes in multivariate analyses. Our data suggest that certain HLA factors may be more significant in some cases and should be given priority over simply selecting a donor based on relationship/sex.
Collapse
Affiliation(s)
- Rohtesh S Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Kai Cao
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rima M Saliba
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Konstantinos Lontos
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Curtis Marcoux
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yudith Carmazzi
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chitra M Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Uday R Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
10
|
Kacen A, Javitt A, Kramer MP, Morgenstern D, Tsaban T, Shmueli MD, Teo GC, da Veiga Leprevost F, Barnea E, Yu F, Admon A, Eisenbach L, Samuels Y, Schueler-Furman O, Levin Y, Nesvizhskii AI, Merbl Y. Post-translational modifications reshape the antigenic landscape of the MHC I immunopeptidome in tumors. Nat Biotechnol 2023; 41:239-251. [PMID: 36203013 PMCID: PMC11197725 DOI: 10.1038/s41587-022-01464-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
Post-translational modification (PTM) of antigens provides an additional source of specificities targeted by immune responses to tumors or pathogens, but identifying antigen PTMs and assessing their role in shaping the immunopeptidome is challenging. Here we describe the Protein Modification Integrated Search Engine (PROMISE), an antigen discovery pipeline that enables the analysis of 29 different PTM combinations from multiple clinical cohorts and cell lines. We expanded the antigen landscape, uncovering human leukocyte antigen class I binding motifs defined by specific PTMs with haplotype-specific binding preferences and revealing disease-specific modified targets, including thousands of new cancer-specific antigens that can be shared between patients and across cancer types. Furthermore, we uncovered a subset of modified peptides that are specific to cancer tissue and driven by post-translational changes that occurred in the tumor proteome. Our findings highlight principles of PTM-driven antigenicity, which may have broad implications for T cell-mediated therapies in cancer and beyond.
Collapse
Affiliation(s)
- Assaf Kacen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Javitt
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias P Kramer
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Tsaban
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Merav D Shmueli
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Eilon Barnea
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Arie Admon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lea Eisenbach
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yishai Levin
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yifat Merbl
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
11
|
Abstract
Inflammation is a biological process that dynamically alters the surrounding microenvironment, including participating immune cells. As a well-protected organ surrounded by specialized barriers and with immune privilege properties, the central nervous system (CNS) tightly regulates immune responses. Yet in neuroinflammatory conditions, pathogenic immunity can disrupt CNS structure and function. T cells in particular play a key role in promoting and restricting neuroinflammatory responses, while the inflamed CNS microenvironment can influence and reshape T cell function and identity. Still, the contraction of aberrant T cell responses within the CNS is not well understood. Using autoimmunity as a model, here we address the contribution of CD4 T helper (Th) cell subsets in promoting neuropathology and disease. To address the mechanisms antagonizing neuroinflammation, we focus on the control of the immune response by regulatory T cells (Tregs) and describe the counteracting processes that preserve their identity under inflammatory challenges. Finally, given the influence of the local microenvironment on immune regulation, we address how CNS-intrinsic signals reshape T cell function to mitigate abnormal immune T cell responses.
Collapse
Affiliation(s)
- Nail Benallegue
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Hania Kebir
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jorge I. Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
12
|
Abed A, Law N, Calapre L, Lo J, Bhat V, Bowyer S, Millward M, Gray ES. Human leucocyte antigen genotype association with the development of immune-related adverse events in patients with non-small cell lung cancer treated with single agent immunotherapy. Eur J Cancer 2022; 172:98-106. [PMID: 35759816 DOI: 10.1016/j.ejca.2022.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/23/2022] [Accepted: 05/17/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Biomarkers that predict the risk of immune-mediated adverse events (irAEs) among patients with non-small cell lung cancer (NSCLC) may reduce morbidity and mortality associated with these treatments. METHODS We carried out high resolution human leucocyte antigen (HLA)-I typing on 179 patients with NSCLC treated with anti-program death (PD)-1/program death ligand (PDL)-1. Toxicity data were collected and graded as per common terminology criteria for adverse event (CTCAE) v5.0. We used 14.8-week for landmark analysis to address lead-time bias to investigate the correlation between HLA-I/II zygosity, supertypes and alleles with irAE. Furthermore, we assessed the association for irAE with clinical benefit rate (CBR), progression-free survival (PFS) and overall survival (OS). RESULTS Homozygosity at one or more HLA-I loci, but not HLA-II, was associated with a reduced risk of irAE (relative risk (RR) = 0.61, 95% CI 0.33-0.95, P = 0.035) especially pneumonitis or any grade 3 toxicity. Patients with HLA-A03 supertype had a higher risk of developing irAE (RR = 1.42, 95% CI 1.02-2.01, P = 0.039). The occurrence of any irAE was significantly associated with improved CBR (RR = 1.48, P < 0.0001), PFS (HR = 0.45, P = 0.0003) and OS (HR = 0.34, P < 0.0001). CONCLUSIONS Homozygosity at one or more HLA-I loci may serve as biomarker to predict patients who are unlikely to experience severe irAEs among patients with NSCLC and treated with anti-PD1/PDL1, but less likely to derive clinical benefit. Patients with HLA-I homozygous might benefit from additional therapy.
Collapse
Affiliation(s)
- Afaf Abed
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia; Linear Clinical Research, Nedlands, WA, Australia; School of Medicine, University of Western Australia, Nedlands, Australia.
| | - Ngie Law
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.
| | - Leslie Calapre
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.
| | - Johnny Lo
- School of Sciences, Edith Cowan University, Joondalup, WA, Australia; Centre for Artificial Intelligence and Machine Learning, Edith Cowan University, Joondalup, WA, Australia.
| | - Vikas Bhat
- School of Medicine, University of Western Australia, Nedlands, Australia.
| | - Samantha Bowyer
- Linear Clinical Research, Nedlands, WA, Australia; School of Medicine, University of Western Australia, Nedlands, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.
| | - Michael Millward
- Linear Clinical Research, Nedlands, WA, Australia; School of Medicine, University of Western Australia, Nedlands, Australia.
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.
| |
Collapse
|
13
|
Tomas-Ojer P, Puthenparampil M, Cruciani C, Docampo MJ, Martin R, Sospedra M. Characterization of Antigen-Induced CD4+ T-Cell Senescence in Multiple Sclerosis. Front Neurol 2022; 13:790884. [PMID: 35185762 PMCID: PMC8852676 DOI: 10.3389/fneur.2022.790884] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/04/2022] [Indexed: 12/21/2022] Open
Abstract
Antigen-induced T-cell exhaustion and T-cell senescence are peripheral regulatory mechanisms that control effector T-cell responses. Markers of exhaustion and senescence on T Cells indicate the previous activation by repetitive stimulation with specific antigens. Malignant tumors are accompanied by enhanced T-cell exhaustion and T-cell senescence resulting in immune evasion, while these control mechanisms might be diminished in autoimmune diseases including multiple sclerosis (MS). To better understand the involvement of antigen-induced T-cell senescence in controlling CD4+ T-cell-mediated autoimmune responses in MS, we have analyzed the re-expression of CD45RA and the downregulation of CD28 and CD27 molecules as markers of antigen-induced T-cell senescence in fresh cerebrospinal fluid (CSF)-infiltrating and paired circulating T cells from patients with MS. Patients with different levels of CD4+ T-cell senescence were identified and characterized regarding demographical and clinical features as well as intrathecal markers of neurodegeneration. CD4+ T-cell senescence was also analyzed in control patients to explore a putative deficit of this regulatory mechanism in MS. This study shows heterogeneity of markers of CD4+ T-cell senescence in patients with MS. Patients with high levels of CD4+ T-cell senescence in peripheral blood showed increased frequencies of CSF-infiltrating CD28+ CD27-EM CD4+ T cells with a proinflammatory Th1 functional phenotype. The correlation of these cells with the intrathecal levels of neurofilament light chain, a marker of neurodegeneration, suggests their relevance in disease pathogenesis and the involvement of T-cell senescence in their regulation. Markers of antigen-induced T-senescence, therefore, show promise as a tool to identify pathogenic CD4+ T cells in patients with MS.
Collapse
Affiliation(s)
- Paula Tomas-Ojer
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
| | - Marco Puthenparampil
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
- Department of Neuroscience DNS, University-Hospital of Padova, Padova, Italy
| | - Carolina Cruciani
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
| | - María José Docampo
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research (NIMS), Department of Neurology, University Hospital and University Zurich, Zurich, Switzerland
- *Correspondence: Mireia Sospedra
| |
Collapse
|
14
|
Murúa SR, Farez MF, Quintana FJ. The Immune Response in Multiple Sclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:121-139. [PMID: 34606377 DOI: 10.1146/annurev-pathol-052920-040318] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune, inflammatory, and neurodegenerative disease that affects the central nervous system (CNS). MS is characterized by immune dysregulation, which results in the infiltration of the CNS by immune cells, triggering demyelination, axonal damage, and neurodegeneration. Although the exact causes of MS are not fully understood, genetic and environmental factors are thought to control MS onset and progression. In this article, we review the main immunological mechanisms involved in MS pathogenesis. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sofía Rodríguez Murúa
- Center for Research on Neuroimmunological Diseases (CIEN), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires 1428, Argentina;
| | - Mauricio F Farez
- Center for Research on Neuroimmunological Diseases (CIEN), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires 1428, Argentina;
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
15
|
Cruciani C, Puthenparampil M, Tomas-Ojer P, Jelcic I, Docampo MJ, Planas R, Manogaran P, Opfer R, Wicki C, Reindl M, Jelcic I, Lutterotti A, Martin R, Sospedra M. T-Cell Specificity Influences Disease Heterogeneity in Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1075. [PMID: 34535569 PMCID: PMC8453544 DOI: 10.1212/nxi.0000000000001075] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES Encouraged by the enormous progress that the identification of specific autoantigens added to the understanding of neurologic autoimmune diseases, we undertook here an in-depth study of T-cell specificities in the autoimmune disease multiple sclerosis (MS), for which the spectrum of responsible autoantigens is not fully defined yet. The identification of target antigens in MS is crucial for therapeutic strategies aimed to induce antigen-specific tolerance. In addition, knowledge of relevant T-cell targets can improve our understanding of disease heterogeneity, a hallmark of MS that complicates clinical management. METHODS The proliferative response and interferon gamma (IFN-γ) release of CSF-infiltrating CD4+ T cells from patients with MS against several autoantigens was used to identify patients with different intrathecal T-cell specificities. Fresh CSF-infiltrating and paired circulating lymphocytes in these patients were characterized in depth by ex vivo immunophenotyping and transcriptome analysis of relevant T-cell subsets. Further examination of these patients included CSF markers of inflammation and neurodegeneration and a detailed characterization with respect to demographic, clinical, and MRI features. RESULTS By testing CSF-infiltrating CD4+ T cells from 105 patients with MS against seven long-known myelin and five recently described GDP-l-fucose synthase peptides, we identified GDP-l-fucose synthase and myelin oligodendrocyte glycoprotein (35-55) responder patients. Immunophenotyping of CSF and paired blood samples in these patients revealed a significant expansion of an effector memory (CCR7- CD45RA-) CD27- Th1 CD4+ cell subset in GDP-l-fucose synthase responders. Subsequent transcriptome analysis of this subset demonstrated expression of Th1 and cytotoxicity-associated genes. Patients with different intrathecal T-cell specificities also differ regarding inflammation- and neurodegeneration-associated biomarkers, imaging findings, expression of HLA class II alleles, and seasonal distribution of the time of the lumbar puncture. DISCUSSION Our observations reveal an association between autoantigen reactivity and features of disease heterogeneity that strongly supports an important role of T-cell specificity in MS pathogenesis. These data have the potential to improve patient classification in clinical practice and to guide the development of antigen-specific tolerization strategies.
Collapse
Affiliation(s)
- Carolina Cruciani
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Marco Puthenparampil
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Paula Tomas-Ojer
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Ivan Jelcic
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Maria Jose Docampo
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Raquel Planas
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Praveena Manogaran
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Roland Opfer
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Carla Wicki
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Markus Reindl
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Ilijas Jelcic
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Andreas Lutterotti
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Roland Martin
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria
| | - Mireia Sospedra
- From the Neuroimmunology and MS Research (NIMS) (C.C., M.P., P.T.O., I.J., M.J.D., R.P., P.M., C.W., I.J., A.L., R.M., M.S.), Department of Neurology, University Hospital and University Zurich, Switzerland; Department of Neuroscience DNS (M.P.), University-Hospital of Padova, Italy; Jung Diagnostics GmbH (R.O.), HIP - Health Innovation Port, Germany; Department of Health Sciences and Technology (C.W.), ETH Zurich, Switzerland; and Clinical Department of Neurology (M.R.), Medical University of Innsbruck, Austria.
| |
Collapse
|
16
|
Veroni C, Aloisi F. The CD8 T Cell-Epstein-Barr Virus-B Cell Trialogue: A Central Issue in Multiple Sclerosis Pathogenesis. Front Immunol 2021; 12:665718. [PMID: 34305896 PMCID: PMC8292956 DOI: 10.3389/fimmu.2021.665718] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The cause and the pathogenic mechanisms leading to multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), are still under scrutiny. During the last decade, awareness has increased that multiple genetic and environmental factors act in concert to modulate MS risk. Likewise, the landscape of cells of the adaptive immune system that are believed to play a role in MS immunopathogenesis has expanded by including not only CD4 T helper cells but also cytotoxic CD8 T cells and B cells. Once the key cellular players are identified, the main challenge is to define precisely how they act and interact to induce neuroinflammation and the neurodegenerative cascade in MS. CD8 T cells have been implicated in MS pathogenesis since the 80's when it was shown that CD8 T cells predominate in MS brain lesions. Interest in the role of CD8 T cells in MS was revived in 2000 and the years thereafter by studies showing that CNS-recruited CD8 T cells are clonally expanded and have a memory effector phenotype indicating in situ antigen-driven reactivation. The association of certain MHC class I alleles with MS genetic risk implicates CD8 T cells in disease pathogenesis. Moreover, experimental studies have highlighted the detrimental effects of CD8 T cell activation on neural cells. While the antigens responsible for T cell recruitment and activation in the CNS remain elusive, the high efficacy of B-cell depleting drugs in MS and a growing number of studies implicate B cells and Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus that is strongly associated with MS, in the activation of pathogenic T cells. This article reviews the results of human studies that have contributed to elucidate the role of CD8 T cells in MS immunopathogenesis, and discusses them in light of current understanding of autoreactivity, B-cell and EBV involvement in MS, and mechanism of action of different MS treatments. Based on the available evidences, an immunopathological model of MS is proposed that entails a persistent EBV infection of CNS-infiltrating B cells as the target of a dysregulated cytotoxic CD8 T cell response causing CNS tissue damage.
Collapse
Affiliation(s)
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
17
|
Wu J, Engdahl E, Gustafsson R, Fogdell-Hahn A, Waterboer T, Hillert J, Olsson T, Alfredsson L, Hedström AK. High antibody levels against human herpesvirus-6A interact with lifestyle factors in multiple sclerosis development. Mult Scler 2021; 28:383-392. [PMID: 34124961 DOI: 10.1177/13524585211022011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Infection with human herpesvirus 6A (HHV-6A) has been suggested to increase multiple sclerosis (MS) risk. However, potential interactions between HHV-6A and environmental/lifestyle risk factors for MS have not previously been studied. METHODS We used two Swedish population-based case-control studies comprising 5993 cases and 5995 controls. Using logistic regression models, subjects with different HHV-6A antibody levels, environmental exposures, and lifestyle habits were compared regarding MS risk, by calculating odds ratios (ORs) with 95% confidence intervals (CIs). Potential interactions between high HHV-6A antibody levels and common environmental exposures and lifestyle factors were evaluated on the additive scale. RESULTS High HHV-6A antibody levels were associated with increased risk of developing MS (OR = 1.5, 95% CI = 1.4-1.6). Regarding MS risk, significant interactions were observed between high HHV-6A antibody levels and both smoking (attributable proportion (AP) = 0.2, 95% CI = 0.1-0.3), low ultraviolet radiation (UVR) exposure (AP = 0.3, 95% CI = 0.1-0.4), and low vitamin D levels (AP = 0.3, 95% CI = 0.0-0.6). CONCLUSION High HHV-6A antibody levels are associated with increased MS risk and act synergistically with common environmental/lifestyle risk factors for MS. Further research is needed to investigate potential mechanisms underlying the interactions presented in this study.
Collapse
Affiliation(s)
- Jing Wu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden/Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Elin Engdahl
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rasmus Gustafsson
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tim Waterboer
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden/Department of Research and Education, Karolinska University Hospital, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Karin Hedström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Identification of two novel bullous pemphigoid- associated alleles, HLA-DQA1*05:05 and -DRB1*07:01, in Germans. Orphanet J Rare Dis 2021; 16:228. [PMID: 34011352 PMCID: PMC8136166 DOI: 10.1186/s13023-021-01863-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/10/2021] [Indexed: 11/17/2022] Open
Abstract
Bullous pemphigoid (BP) is the most common autoimmune skin blistering disease characterized by autoimmunity against the hemidesmosomal proteins BP180, type XVII collagen, and BP230. To elucidate the genetic basis of susceptibility to BP, we performed the first genome-wide association study (GWAS) in Germans. This GWAS was combined with HLA locus targeted sequencing in an additional independent BP cohort. The strongest association with BP in Germans tested in this study was observed in the two HLA loci, HLA-DQA1*05:05 and HLA-DRB1*07:01. Further studies with increased sample sizes and complex studies integrating multiple pathogenic drivers will be conducted.
Collapse
|
19
|
Maia A, Barahona-Corrêa B, Oliveira-Maia AJ, Oliveira J. Immune Dysfunction in Obsessive-Compulsive Disorder: From Risk Factors to Multisystem Involvement. IMMUNO-PSYCHIATRY 2021:289-307. [DOI: 10.1007/978-3-030-71229-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Houen G, Trier NH, Frederiksen JL. Epstein-Barr Virus and Multiple Sclerosis. Front Immunol 2020; 11:587078. [PMID: 33391262 PMCID: PMC7773893 DOI: 10.3389/fimmu.2020.587078] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neurologic disease affecting myelinated nerves in the central nervous system (CNS). The disease often debuts as a clinically isolated syndrome, e.g., optic neuritis (ON), which later develops into relapsing-remitting (RR) MS, with temporal attacks or primary progressive (PP) MS. Characteristic features of MS are inflammatory foci in the CNS and intrathecal synthesis of immunoglobulins (Igs), measured as an IgG index, oligoclonal bands (OCBs), or specific antibody indexes. Major predisposing factors for MS are certain tissue types (e.g., HLA DRB1*15:01), vitamin D deficiency, smoking, obesity, and infection with Epstein-Barr virus (EBV). Many of the clinical signs of MS described above can be explained by chronic/recurrent EBV infection and current models of EBV involvement suggest that RRMS may be caused by repeated entry of EBV-transformed B cells to the CNS in connection with attacks, while PPMS may be caused by more chronic activity of EBV-transformed B cells in the CNS. In line with the model of EBV's role in MS, new treatments based on monoclonal antibodies (MAbs) targeting B cells have shown good efficacy in clinical trials both for RRMS and PPMS, while MAbs inhibiting B cell mobilization and entry to the CNS have shown efficacy in RRMS. Thus, these agents, which are now first line therapy in many patients, may be hypothesized to function by counteracting a chronic EBV infection.
Collapse
Affiliation(s)
- Gunnar Houen
- Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | | | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet, Glostrup, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Huang S, Zheng F, Liu L, Meng S, Cai W, Zhang C, Dai W, Liu D, Hong X, Tang D, Dai Y. Integrated proteome and phosphoproteome analyses of peripheral blood mononuclear cells in primary Sjögren syndrome patients. Aging (Albany NY) 2020; 13:1071-1095. [PMID: 33290261 PMCID: PMC7835054 DOI: 10.18632/aging.202233] [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: 06/18/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022]
Abstract
Primary Sjögren syndrome (pSS) is a common autoimmune disease. Here, we performed the first proteome and phosphoproteome analyses of peripheral blood mononuclear cells in pSS patients to obtain a comprehensive profile and identify the potential crucial proteins and pathways for the screening and evaluation of pSS patients. Peripheral blood mononuclear cells from 8 pSS-confirmed patients (American-European Consensus Group Criteria, 2002) and 10 normal controls were selected. Label-free quantitative proteomics was utilized to obtain quantitative information. In total, 787 proteins were identified as differentially expressed proteins, and 175 phosphosites on 123 proteins were identified as differentially phosphorylated proteins. We performed functional enrichment analyses with these proteins and phosphoproteins based on public database. Furthermore, protein-protein interaction network analyses were performed by using multiple algorithms. Using module and hub protein analyses, we identified 16 modules for the proteins, 2 clusters for the phosphoproteins and selected the top 10 hub proteins. Finally, we identified 22 motifs using motif analysis of the phosphosites and found 17 newly identified motifs, while 6 motifs were experimentally verified for known protein kinases. The findings distinguished pSS patients from normal controls at the peripheral blood mononuclear cells level and revealed potential candidates for use in pSS diagnosis.
Collapse
Affiliation(s)
- Shaoying Huang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Fengping Zheng
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Lixiong Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Shuhui Meng
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Wanxia Cai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Cantong Zhang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, Guangdong, China
| | - Donge Tang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | - Yong Dai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen People’s Hospital, The First Affiliated Hospital Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China.,Guangxi Key Laboratory of Metabolic Disease Research, Nephrology Department of Guilin, Guilin 541002, China
| |
Collapse
|
22
|
Ferrè L, Filippi M, Esposito F. Involvement of Genetic Factors in Multiple Sclerosis. Front Cell Neurosci 2020; 14:612953. [PMID: 33335478 PMCID: PMC7735985 DOI: 10.3389/fncel.2020.612953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/09/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Laura Ferrè
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurophysiology Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Federica Esposito
- Neurology Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
24
|
Zhao X, Li J, Zhu Q, Liang G, Xia W, He X, Zhu C, Qi H, Deng B, Chen X, Zhang J. HLA-A and HLA-DRB1 may play a unique role in ovarian teratoma-associated anti-N-methyl-D-aspartate receptor encephalitis. Reprod Biol Endocrinol 2020; 18:107. [PMID: 33160385 PMCID: PMC7648266 DOI: 10.1186/s12958-020-00661-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian teratoma-associated anti-N-methyl-D-aspartate receptor encephalitis (NMDAR-E) is a severe autoimmune neurological disorder, and the influence of teratoma-induced autoantibodies on the pathogenesis remains unclear. METHODS Ovarian teratoma tissues were collected from teratoma patients with and without NMDAR-E. Proteins were extracted and then analyzed using iTRAQ-coupled LC-MS/MS, which was followed by bioinformatics analysis. Candidate proteins were verified by Western blotting and immunohistochemistry. RESULTS In total, 36 differentially expressed proteins (DEPs) were identified between the control group and NMDAR-E group, and the bioinformatics analysis revealed that the DEPs were mainly involved in immune-related pathways, especially HLA-A and HLA-DRB1. The western blotting results for HLA-A and HLA-DRB1 were consistent with the results of the iTRAQ analysis. Additionally, the immunohistochemical data revealed that the aggregation of HLA-A (+) and HLA-DRB1 (+) cells was more apparent in the teratoma tissues of NMDAR-E patients compared with that in the tissues of controls. CONCLUSION Our investigation indicated that HLA-A and HLA-DRB1 might be involved in mediating ovarian teratoma-associated NMDAR-E. These findings provide new insights into the pathophysiological mechanisms and provide information for the functional exploration of proteins in the future.
Collapse
Affiliation(s)
- Xiaoya Zhao
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Juan Li
- Department of Pathology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Qian Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Guiling Liang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Wei Xia
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Xiaoqing He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Chenfeng Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Hang Qi
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China
| | - Bo Deng
- Department of Neurology, Huashan Hospital and Institute of Neurology, Fudan University, No. 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, Fudan University, No. 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| | - Jian Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, No. 910, Hengshan Rd, Shanghai, 200030, China.
| |
Collapse
|
25
|
Mescheriakova JY, van Nierop GP, van der Eijk AA, Kreft KL, Hintzen RQ. EBNA-1 titer gradient in families with multiple sclerosis indicates a genetic contribution. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/6/e872. [PMID: 32796079 PMCID: PMC7428359 DOI: 10.1212/nxi.0000000000000872] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE In multiplex MS families, we determined the humoral immune response to Epstein-Barr virus nuclear antigen 1 (EBNA-1)-specific immunoglobulin γ (IgG) titers in patients with MS, their healthy siblings, and biologically unrelated healthy spouses and investigated the role of specific genetic loci on the antiviral IgG titers. METHODS IgG levels against EBNA-1 and varicella zoster virus (VZV) as control were measured. HLA-DRB1*1501 and HLA-A*02 tagging single-nucleotide polymorphisms (SNPs) were genotyped. We assessed the associations between these SNPs and antiviral IgG titers. RESULTS OR for abundant EBNA-1 IgG was the highest in patients with MS and intermediate in their siblings compared with spouses. We confirmed that HLA-DRB1*1501 is associated with abundant EBNA-1 IgG. After stratification for HLA-DRB1*1501, the EBNA-1 IgG gradient was still significant in patients with MS and young siblings compared with spouses. HLA-A*02 was not explanatory for EBNA-1 IgG titer gradient. No associations for VZV IgG were found. CONCLUSIONS In families with MS, the EBNA-1 IgG gradient being the highest in patients with MS, intermediate in their siblings, and lowest in biologically unrelated spouses indicates a genetic contribution to EBNA-1 IgG levels that is only partially explained by HLA-DRB1*1501 carriership.
Collapse
Affiliation(s)
- Julia Y Mescheriakova
- From the Department of Neurology (J.Y.M., K.L.K.); Department of Viroscience (G.P.N., A.A.E.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; and Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands (J.Y.M., K.L.K.)
| | - Gijsbert P van Nierop
- From the Department of Neurology (J.Y.M., K.L.K.); Department of Viroscience (G.P.N., A.A.E.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; and Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands (J.Y.M., K.L.K.)
| | - Annemiek A van der Eijk
- From the Department of Neurology (J.Y.M., K.L.K.); Department of Viroscience (G.P.N., A.A.E.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; and Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands (J.Y.M., K.L.K.)
| | - Karim L Kreft
- From the Department of Neurology (J.Y.M., K.L.K.); Department of Viroscience (G.P.N., A.A.E.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; and Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands (J.Y.M., K.L.K.).
| | - Rogier Q Hintzen
- From the Department of Neurology (J.Y.M., K.L.K.); Department of Viroscience (G.P.N., A.A.E.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; and Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands (J.Y.M., K.L.K.).
| |
Collapse
|
26
|
Abstract
Multiple sclerosis (MS) is an aggravating autoimmune disease that cripples young patients slowly with physical, sensory and cognitive deficits. The break of self-tolerance to neuronal antigens is the key to the pathogenesis of MS, with autoreactive T cells causing demyelination that subsequently leads to inflammation-mediated neurodegenerative events in the central nervous system. The exact etiology of MS remains elusive; however, the interplay of genetic and environmental factors contributes to disease development and progression. Given that genetic variation only accounts for a fraction of risk for MS, extrinsic risk factors including smoking, infection and lack of vitamin D or sunshine, which cause changes in gene expression, contribute to disease development through epigenetic regulation. To date, there is a growing body of scientific evidence to support the important roles of epigenetic processes in MS. In this chapter, the three main layers of epigenetic regulatory mechanisms, namely DNA methylation, histone modification and microRNA-mediated gene regulation, will be discussed, with a particular focus on the role of epigenetics on dysregulated immune responses and neurodegenerative events in MS. Also, the potential for epigenetic modifiers as biomarkers and therapeutics for MS will be reviewed.
Collapse
Affiliation(s)
- Vera Sau-Fong Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Queen Mary Hospital, Hong Kong SAR, China.
| |
Collapse
|
27
|
Batal I, Vasilescu ER, Dadhania DM, Adel AA, Husain SA, Avasare R, Serban G, Santoriello D, Khairallah P, Patel A, Moritz MJ, Latulippe E, Riopel J, Khallout K, Swanson SJ, Bomback AS, Mohan S, Ratner L, Radhakrishnan J, Cohen DJ, Appel GB, Stokes MB, Markowitz GS, Seshan SV, De Serres SA, Andeen N, Loupy A, Kiryluk K, D'Agati VD. Association of HLA Typing and Alloimmunity With Posttransplantation Membranous Nephropathy: A Multicenter Case Series. Am J Kidney Dis 2020; 76:374-383. [PMID: 32359820 PMCID: PMC7483441 DOI: 10.1053/j.ajkd.2020.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE & OBJECTIVES Posttransplantation membranous nephropathy (MN) represents a rare complication of kidney transplantation that can be classified as recurrent or de novo. The clinical, pathologic, and immunogenetic characteristics of posttransplantation MN and the differences between de novo and recurrent MN are not well understood. STUDY DESIGN Multicenter case series. SETTING & PARTICIPANTS We included 77 patients from 5 North American and European medical centers with post-kidney transplantation MN (27 de novo and 50 recurrent). Patients with MN in the native kidney who received kidney allografts but did not develop recurrent MN were used as nonrecurrent controls (n = 43). To improve understanding of posttransplantation MN, we compared de novo MN with recurrent MN and then contrasted recurrent MN with nonrecurrent controls. FINDINGS Compared with recurrent MN, de novo MN was less likely to be classified as primary MN (OR, 0.04; P < 0.001) and had more concurrent antibody-mediated rejection (OR, 12.0; P < 0.001) and inferior allograft survival (HR for allograft failure, 3.2; P = 0.007). HLA-DQ2 and HLA-DR17 antigens were more common in recipients with recurrent MN compared with those with de novo MN; however, the frequency of these recipient antigens in recurrent MN was similar to that in nonrecurrent MN controls. Among the 93 kidney transplant recipients with native kidney failure attributed to MN, older recipient age (HR per each year older, 1.03; P = 0.02), recipient HLA-A3 antigen (HR, 2.5; P = 0.003), steroid-free immunosuppressive regimens (HR, 2.84; P < 0.001), and living related allograft (HR, 1.94; P = 0.03) were predictors of MN recurrence. LIMITATIONS Retrospective case series, limited sample size due to rarity of the disease, nonstandardized nature of data collection and biopsies. CONCLUSIONS De novo and recurrent MN likely represent separate diseases. De novo MN is associated with humoral alloimmunity and guarded outcome. Potential predisposing factors for recurrent MN include recipients who are older, recipient HLA-A3 antigen, steroid-free immunosuppressive regimen, and living related donor kidney.
Collapse
Affiliation(s)
- Ibrahim Batal
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY.
| | - Elena-Rodica Vasilescu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Darshana M Dadhania
- Department of Medicine, Nephrology, Weill Cornell Medical College, New York, NY
| | | | - S Ali Husain
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Rupali Avasare
- Department of Medicine, Nephrology, Oregon Health & Science University, Portland, OR
| | - Geo Serban
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Dominick Santoriello
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Pascale Khairallah
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Ankita Patel
- Department of Medicine, Nephrology, Hackensack University Medical Center, Hackensack, NJ
| | - Michael J Moritz
- Department of Surgery, Lehigh Valley Health Network, Allentown, PA
| | - Eva Latulippe
- Department of Pathology, University Health Center of Quebec, Laval University, Québec, QC, Canada
| | - Julie Riopel
- Department of Pathology, University Health Center of Quebec, Laval University, Québec, QC, Canada
| | - Karim Khallout
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | | | - Andrew S Bomback
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Sumit Mohan
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Lloyd Ratner
- Department of Surgery, Columbia University Irving Medical Center, New York, NY
| | - Jai Radhakrishnan
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - David J Cohen
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Gerald B Appel
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Michael B Stokes
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Glen S Markowitz
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Surya V Seshan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Sacha A De Serres
- Renal Division, Department of Medicine, University Health Center of Quebec, Laval University, Québec, QC, Canada
| | - Nicole Andeen
- Department of Pathology, Oregon Health & Science University, Portland, OR
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Krzysztof Kiryluk
- Department of Medicine, Nephrology, Columbia University Irving Medical Center, New York, NY
| | - Vivette D D'Agati
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
28
|
Stürner KH, Siembab I, Schön G, Stellmann JP, Heidari N, Fehse B, Heesen C, Eiermann TH, Martin R, Binder TM. Is multiple sclerosis progression associated with the HLA-DR15 haplotype? Mult Scler J Exp Transl Clin 2019; 5:2055217319894615. [PMID: 31839982 PMCID: PMC6902395 DOI: 10.1177/2055217319894615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/04/2019] [Accepted: 11/09/2019] [Indexed: 11/17/2022] Open
Abstract
Background The prevalence of multiple sclerosis is associated with the major histocompatibility complex class II DR15 haplotype HLA-DRB1*15:01∼HLA-DRB5*01:01. Objective To assess whether multiple sclerosis progression is associated with the main susceptibility haplotype HLA-DRB1*15:01∼HLA-DRB5*01:01. Methods Patients (n = 1230) and healthy controls (n = 2110) were genotyped for HLA-DRB1 and HLA-DRB5. The baseline Expanded Disability Status Scale (EDSS) score was determined and patients were followed for at least 3 years. Results After follow-up of the consecutive cohort 349 patients were classified as having clinical isolated syndrome and 881 patients as having multiple sclerosis. The susceptibility allele HLA-DRB1*15:01 was more frequent in clinical isolated syndrome (odds ratio 1.56) and multiple sclerosis (odds ratio 3.17) compared to controls. HLA- DRB1*15:01 was the only enriched HLA-DRB1 allele in multiple sclerosis patients. Comparison of clinical characteristics between HLA-DRB1*15:01∼HLA-DRB5*01:01 negative and positive patients with multiple sclerosis showed that baseline EDSS score, disease duration and frequency of the category secondary progressive multiple sclerosis with relapse were increased in the HLA-DRB1*15:01∼HLA-DRB5*01:01 positive group. Conclusion The study confirmed HLA-DRB1*15:01 and HLA-DRB5*01:01 as the main susceptibility alleles and showed weak indirect evidence for a role in progression of the disease.
Collapse
Affiliation(s)
- Klarissa Hanja Stürner
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Inessa Siembab
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Gerhard Schön
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Germany
| | - Jan-Patrick Stellmann
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Nika Heidari
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Germany
| | - Christoph Heesen
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Thomas H Eiermann
- HLA Laboratory, University Medical Center Hamburg-Eppendorf, Germany
| | - Roland Martin
- Institute for Neuroimmunology and Clinical MS Research, University Medical Center Hamburg-Eppendorf, Germany
| | - Thomas Mc Binder
- HLA Laboratory, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
29
|
Abstract
OBJECTIVE In a previous pilot monocentric study, we investigated the relation between human leukocyte antigen (HLA) genotype and multiple sclerosis (MS) disease progression over 2 years. HLA-A*02 allele was correlated with better outcomes, whereas HLA-B*07 and HLA-B*44 were correlated with worse outcomes. The objective of this extension study was to further investigate the possible association of HLA genotype with disease status and progression in MS as measured by sensitive and complex clinical and imaging parameters. METHODS Hundred and forty-six MS patients underwent HLA typing. Over a 4-year period of follow-up, we performed three clinical and magnetic resonance imaging (MRI) assessments per patient, which respectively included Expanded Disability Status Scale, Multiple Sclerosis Severity Scale, Timed-25-Foot-Walk, 9-Hole Peg Test, Symbol Digit Modalities Test, Brief Visual Memory Test, California Verbal Learning Test-II, and whole-brain atrophy, fluid-attenuated inversion recovery (FLAIR) lesion volume change and number of new FLAIR lesions using icobrain. We then compared the clinical and MRI outcomes between predefined HLA patient groups. RESULTS Results of this larger study with a longer follow-up are in line with what we have previously shown. HLA-A*02 allele is associated with potentially better MS outcomes, whereas HLA-B*07, HLA-B*44, HLA-B*08, and HLA-DQB1*06 with a potential negative effect. Results for HLA-DRB1*15 are inconclusive. CONCLUSION In the era of MS treatment abundance, HLA genotype might serve as an early biomarker for MS outcomes to inform individualized treatment decisions.
Collapse
|
30
|
Anti-CD20 therapy depletes activated myelin-specific CD8 + T cells in multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:25800-25807. [PMID: 31748274 PMCID: PMC6926057 DOI: 10.1073/pnas.1915309116] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. CD8+ T cells have been strongly implicated in MS pathogenesis, but it is unclear whether myelin is a CD8+ T cell autoantigenic target in MS. This study demonstrated that while myelin-specific CD8+ T cells are present at similar frequencies in untreated MS patients and healthy subjects, the proportion of memory and CD20-expressing myelin-specific CD8+ T cells was increased in MS patients, suggesting prior antigen encounter. This activated phenotype was reversible as the memory and CD20-expressing populations of certain myelin-specific CD8+ T cells were reduced following anti-CD20 treatment. CD8+ T cells are believed to play an important role in multiple sclerosis (MS), yet their role in MS pathogenesis remains poorly defined. Although myelin proteins are considered potential autoantigenic targets, prior studies of myelin-reactive CD8+ T cells in MS have relied on in vitro stimulation, thereby limiting accurate measurement of their ex vivo precursor frequencies and phenotypes. Peptide:MHC I tetramers were used to identify and validate 5 myelin CD8+ T cell epitopes, including 2 newly described determinants in humans. The validated tetramers were used to measure the ex vivo precursor frequencies and phenotypes of myelin-specific CD8+ T cells in the peripheral blood of untreated MS patients and HLA allele-matched healthy controls. In parallel, CD8+ T cell responses against immunodominant influenza epitopes were also measured. There were no differences in ex vivo frequencies of tetramer-positive myelin-specific CD8+ T cells between MS patients and control subjects. An increased proportion of myelin-specific CD8+ T cells in MS patients exhibited a memory phenotype and expressed CD20 compared to control subjects, while there were no phenotypic differences observed among influenza-specific CD8+ T cells. Longitudinal assessments were also measured in a subset of MS patients subsequently treated with anti-CD20 monoclonal antibody therapy. The proportion of memory and CD20+ CD8+ T cells specific for certain myelin but not influenza epitopes was significantly reduced following anti-CD20 treatment. This study, representing a characterization of unmanipulated myelin-reactive CD8+ T cells in MS, indicates these cells may be attractive targets in MS therapy.
Collapse
|
31
|
Planas R, Santos R, Tomas-Ojer P, Cruciani C, Lutterotti A, Faigle W, Schaeren-Wiemers N, Espejo C, Eixarch H, Pinilla C, Martin R, Sospedra M. GDP-l-fucose synthase is a CD4 + T cell-specific autoantigen in DRB3*02:02 patients with multiple sclerosis. Sci Transl Med 2019; 10:10/462/eaat4301. [PMID: 30305453 DOI: 10.1126/scitranslmed.aat4301] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis is an immune-mediated autoimmune disease of the central nervous system that develops in genetically susceptible individuals and likely requires environmental triggers. The autoantigens and molecular mimics triggering the autoimmune response in multiple sclerosis remain incompletely understood. By using a brain-infiltrating CD4+ T cell clone that is clonally expanded in multiple sclerosis brain lesions and a systematic approach for the identification of its target antigens, positional scanning peptide libraries in combination with biometrical analysis, we have identified guanosine diphosphate (GDP)-l-fucose synthase as an autoantigen that is recognized by cerebrospinal fluid-infiltrating CD4+ T cells from HLA-DRB3*-positive patients. Significant associations were found between reactivity to GDP-l-fucose synthase peptides and DRB3*02:02 expression, along with reactivity against an immunodominant myelin basic protein peptide. These results, coupled with the cross-recognition of homologous peptides from gut microbiota, suggest a possible role of this antigen as an inducer or driver of pathogenic autoimmune responses in multiple sclerosis.
Collapse
Affiliation(s)
- Raquel Planas
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Radleigh Santos
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway Port St. Lucie, FL 34987, USA
| | - Paula Tomas-Ojer
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Carolina Cruciani
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Andreas Lutterotti
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Nicole Schaeren-Wiemers
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway Port St. Lucie, FL 34987, USA
| | - Roland Martin
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zürich, Switzerland.
| |
Collapse
|
32
|
Rahmanzadeh R, Brück W, Minagar A, Sahraian MA. Multiple sclerosis pathogenesis: missing pieces of an old puzzle. Rev Neurosci 2019; 30:67-83. [PMID: 29883325 DOI: 10.1515/revneuro-2018-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/30/2018] [Indexed: 11/15/2022]
Abstract
Traditionally, multiple sclerosis (MS) was considered to be a CD4 T cell-mediated CNS autoimmunity, compatible with experimental autoimmune encephalitis model, which can be characterized by focal lesions in the white matter. However, studies of recent decades revealed several missing pieces of MS puzzle and showed that MS pathogenesis is more complex than the traditional view and may include the following: a primary degenerative process (e.g. oligodendroglial pathology), generalized abnormality of normal-appearing brain tissue, pronounced gray matter pathology, involvement of innate immunity, and CD8 T cells and B cells. Here, we review these findings and discuss their implications in MS pathogenesis.
Collapse
Affiliation(s)
- Reza Rahmanzadeh
- MS Research Center, Neuroscience Institute, Tehran University of Medical Science, Department of Neurology, Sina Hospital, 1136746911 Tehran, Iran
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center, D-37075 Göttingen, Germany
| | - Alireza Minagar
- Department of Neurology, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | - Mohammad Ali Sahraian
- MS Research Center, Neuroscience Institute, Tehran University of Medical Science, Department of Neurology, Sina Hospital, 1136746911 Tehran, Iran.,Iranian Center for Neurological Research, Neuroscience Institute, Tehran University of Medical Science, 1136746890 Tehran, Iran
| |
Collapse
|
33
|
Non-parametric combination analysis of multiple data types enables detection of novel regulatory mechanisms in T cells of multiple sclerosis patients. Sci Rep 2019; 9:11996. [PMID: 31427643 PMCID: PMC6700160 DOI: 10.1038/s41598-019-48493-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system with prominent neurodegenerative components. The triggering and progression of MS is associated with transcriptional and epigenetic alterations in several tissues, including peripheral blood. The combined influence of transcriptional and epigenetic changes associated with MS has not been assessed in the same individuals. Here we generated paired transcriptomic (RNA-seq) and DNA methylation (Illumina 450 K array) profiles of CD4+ and CD8+ T cells (CD4, CD8), using clinically accessible blood from healthy donors and MS patients in the initial relapsing-remitting and subsequent secondary-progressive stage. By integrating the output of a differential expression test with a permutation-based non-parametric combination methodology, we identified 149 differentially expressed (DE) genes in both CD4 and CD8 cells collected from MS patients. Moreover, by leveraging the methylation-dependent regulation of gene expression, we identified the gene SH3YL1, which displayed significant correlated expression and methylation changes in MS patients. Importantly, silencing of SH3YL1 in primary human CD4 cells demonstrated its influence on T cell activation. Collectively, our strategy based on paired sampling of several cell-types provides a novel approach to increase sensitivity for identifying shared mechanisms altered in CD4 and CD8 cells of relevance in MS in small sized clinical materials.
Collapse
|
34
|
Molecular mimicry between Anoctamin 2 and Epstein-Barr virus nuclear antigen 1 associates with multiple sclerosis risk. Proc Natl Acad Sci U S A 2019; 116:16955-16960. [PMID: 31375628 DOI: 10.1073/pnas.1902623116] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, likely autoimmune disease of the central nervous system with a combination of genetic and environmental risk factors, among which Epstein-Barr virus (EBV) infection is a strong suspect. We have previously identified increased autoantibody levels toward the chloride-channel protein Anoctamin 2 (ANO2) in MS. Here, IgG antibody reactivity toward ANO2 and EBV nuclear antigen 1 (EBNA1) was measured using bead-based multiplex serology in plasma samples from 8,746 MS cases and 7,228 controls. We detected increased anti-ANO2 antibody levels in MS (P = 3.5 × 10-36) with 14.6% of cases and 7.8% of controls being ANO2 seropositive (odds ratio [OR] = 1.6; 95% confidence intervals [95%CI]: 1.5 to 1.8). The MS risk increase in ANO2-seropositive individuals was dramatic when also exposed to 3 known risk factors for MS: HLA-DRB1*15:01 carriage, absence of HLA-A*02:01, and high anti-EBNA1 antibody levels (OR = 24.9; 95%CI: 17.9 to 34.8). Reciprocal blocking experiments with ANO2 and EBNA1 peptides demonstrated antibody cross-reactivity, mapping to ANO2 [aa 140 to 149] and EBNA1 [aa 431 to 440]. HLA gene region was associated with anti-ANO2 antibody levels and HLA-DRB1*04:01 haplotype was negatively associated with ANO2 seropositivity (OR = 0.6; 95%CI: 0.5 to 0.7). Anti-ANO2 antibody levels were not increased in patients from 3 other inflammatory disease cohorts. The HLA influence and the fact that specific IgG production usually needs T cell help provides indirect evidence for a T cell ANO2 autoreactivity in MS. We propose a hypothesis where immune reactivity toward EBNA1 through molecular mimicry with ANO2 contributes to the etiopathogenesis of MS.
Collapse
|
35
|
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 PMCID: PMC6804597 DOI: 10.1038/s41423-019-0221-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
36
|
Mack SJ, Udell J, Cohen F, Osoegawa K, Hawbecker SK, Noonan DA, Ladner MB, Goodridge D, Trachtenberg EA, Oksenberg JR, Erlich HA. High resolution HLA analysis reveals independent class I haplotypes and amino-acid motifs protective for multiple sclerosis. Genes Immun 2019; 20:308-326. [PMID: 29307888 PMCID: PMC6035897 DOI: 10.1038/s41435-017-0006-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 11/24/2022]
Abstract
We investigated association between HLA class I and class II alleles and haplotypes, and KIR loci and their HLA class I ligands, with multiple sclerosis (MS) in 412 European American MS patients and 419 ethnically matched controls, using next-generation sequencing. The DRB1*15:01~DQB1*06:02 haplotype was highly predisposing (odds ratio (OR) = 3.98; 95% confidence interval (CI) = 3-5.31; p-value (p) = 2.22E-16), as was DRB1*03:01~DQB1*02:01 (OR = 1.63; CI = 1.19-2.24; p = 1.41E-03). Hardy-Weinberg (HW) analysis in MS patients revealed a significant DRB1*03:01~DQB1*02:01 homozyote excess (15 observed; 8.6 expected; p = 0.016). The OR for this genotype (5.27; CI = 1.47-28.52; p = 0.0036) suggests a recessive MS risk model. Controls displayed no HW deviations. The C*03:04~B*40:01 haplotype (OR = 0.27; CI = 0.14-0.51; p = 6.76E-06) was highly protective for MS, especially in haplotypes with A*02:01 (OR = 0.15; CI = 0.04-0.45; p = 6.51E-05). By itself, A*02:01 is moderately protective, (OR = 0.69; CI = 0.54-0.87; p = 1.46E-03), and haplotypes of A*02:01 with the HLA-B Thr80 Bw4 variant (Bw4T) more so (OR = 0.53; CI = 0.35-0.78; p = 7.55E-04). Protective associations with the Bw4 KIR ligand resulted from linkage disequilibrium (LD) with DRB1*15:01, but the Bw4T variant was protective (OR = 0.64; CI = 0.49-0.82; p = 3.37-04) independent of LD with DRB1*15:01. The Bw4I variant was not associated with MS. Overall, we find specific class I HLA polymorphisms to be protective for MS, independent of the strong predisposition conferred by DRB1*15:01.
Collapse
Affiliation(s)
- Steven J Mack
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA.
| | - Julia Udell
- University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Franziska Cohen
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Kazutoyo Osoegawa
- Histocompatibility, Immunogenetics & Disease Profiling Laboratory, Stanford Blood Center, Palo Alto, CA, USA
| | - Sharon K Hawbecker
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - David A Noonan
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Martha B Ladner
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | | | | | - Jorge R Oksenberg
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Henry A Erlich
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| |
Collapse
|
37
|
Li H, Xiao L, Wang L, Lin J, Luo M, Chen M, He R, Zhu Y, Zhang C. HLA Polymorphism Affects Risk of de novo Mutation of dystrophin Gene and Clinical Severity of Duchenne Muscular Dystrophy in a Southern Chinese Population. Front Neurol 2018; 9:970. [PMID: 30498470 PMCID: PMC6249334 DOI: 10.3389/fneur.2018.00970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Immune-mediated pathology has been thought to be an important factor contributing to Duchenne muscular dystrophy (DMD). Allele frequencies of certain HLA types are known to differ between patients with dystrophinopathies and healthy controls with low-resolution HLA gene typing data in limit reports. Using Polymerase chain reactionsequence based typing (PCR-SBT) to genotype 64 children with DMD in HLA-A, -B,-C, -DRB1, and -DQB1 locus and 503 healthy controls in HLA-A, -B, -DRB1 locus, this study aimed to investigate associations of specific HLA alleles with, and their possible roles in the development and clinical phenotypic severity of DMD. The χ2 test was used to evaluate the distribution of allele frequencies in HLA-A, -B, -DRB1 locus between the patients and healthy controls. A significantly higher frequency of HLA-B*07:05 was found in children with DMD compared to that in controls (OR = 16.2, 95%CI = 2.9–89.3, P < 0.046). More importantly, significantly higher frequencies of HLA-A*29:01 (OR = 77.308, 95%CI = 6.794–879.731, P < 0.0160) and HLA-B*07:05 (OR = 60.240, 95%CI = 9.637–376.535, P < 2.41*10−3) was found in patients with de novo mutations (n = 14) compared to controls while no difference of HLA alleles frequency ware indicated between patients with inherited mutation and control. The result indicates that HLA alleles is associated with pathogenesis of DMD especially DMD with de novo mutation. We use Vignos scale to estimate the lower limb motor function of patients. The impact of HLA alleles on score of Vignos scale of DMD children was estimated by multiple linear regression. Our study indicates that HLA-A*02:01 may have a dampening effect on the clinical phenotypic severity of DMD, evidenced by the presence of HLA-A*02:01 being associated with lower Vignos score. Our study demonstrates that certain HLA alleles are indeed associated with the pathogenesis and clinical phenotypic severity of DMD.
Collapse
Affiliation(s)
- Huan Li
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lulu Xiao
- Department of Tissue Typing Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinfu Lin
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Luo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruojie He
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuling Zhu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng Zhang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Latt KZ, Honda K, Thiri M, Hitomi Y, Omae Y, Sawai H, Kawai Y, Teraguchi S, Ueno K, Nagasaki M, Mabuchi A, Kaga H, Komatsuda A, Tokunaga K, Noiri E. Identification of a two-SNP PLA2R1 Haplotype and HLA-DRB1 Alleles as Primary Risk Associations in Idiopathic Membranous Nephropathy. Sci Rep 2018; 8:15576. [PMID: 30349113 PMCID: PMC6197221 DOI: 10.1038/s41598-018-33612-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/03/2018] [Indexed: 01/12/2023] Open
Abstract
The associations of single nucleotide polymorphisms (SNPs) in PLA2R1 and HLA-DQA1, as well as HLA-DRB1*15:01-DQB1*06:02 haplotype with idiopathic membranous nephropathy (IMN) is well known. However, the primary associations of these loci still need to be determined. We used Japanese-specific SNP genotyping array and imputation using 2,048 sequenced Japanese samples to fine-map PLA2R1 region in 98 patients and 413 controls. The most significant SNPs were replicated in a separate sample set of 130 patients and 288 controls. A two-SNP haplotype of intronic and missense SNPs showed the strongest association. The intronic SNP is strongly associated with PLA2R1 expression in the Genotype-Tissue Expression (GTEx) database, and the missense SNP is predicted to alter peptide binding with HLA-DRB1*15:01 by the Immune Epitope Database (IEDB). In HLA region, we performed relative predispositional effect (RPE) tests and identified additional risk alleles in both HLA-DRB1 and HLA-DQB1. We collapsed the risk alleles in each of HLA-DRB1 and HLA-DQB1 into single risk alleles. Reciprocal conditioning of these collapsed risk alleles showed more residual significance for HLA-DRB1 collapsed risk than HLA-DQB1 collapsed risk. These results indicate that changes in the expression levels of structurally different PLA2R protein confer risk for IMN in the presence of risk HLA-DRB1 alleles.
Collapse
Affiliation(s)
- Khun Zaw Latt
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenjiro Honda
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
| | - Myo Thiri
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Hitomi
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Omae
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromi Sawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Kawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Teraguchi
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kazuko Ueno
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Akihiko Mabuchi
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hajime Kaga
- Department of Hematology, Nephrology, and Rheumatology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Atsushi Komatsuda
- Department of Hematology, Nephrology, and Rheumatology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Eisei Noiri
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan. .,Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
39
|
Abstract
Multiple sclerosis (MS) has long been considered a CD4 T-cell disease, primarily because of the findings that the strongest genetic risk for MS is the major histocompatibility complex (MHC) class II locus, and that T cells play a central role in directing the immune response. The importance that the T helper (Th)1 cytokine, interferon γ (IFN-γ), and the Th17 cytokine, interleukin (IL)-17, play in MS pathogenesis is indicated by recent clinical trial data by the enhanced presence of Th1/Th17 cells in central nervous system (CNS) tissue, cerebrospinal fluid (CSF), and blood, and by research on animal models of MS, such as experimental autoimmune encephalomyelitis (EAE). Although the majority of research on MS pathogenesis has centered on the role of effector CD4 T cells, accumulating data suggests that CD8 T cells may play a significant role in the human disease. In fact, in contrast to most animal models, the primary T cell found in the CNS in patients with MS, is the CD8 T cell. As patient-derived effector T cells are also resistant to mechanisms of dominant tolerance such as that induced by interaction with regulatory T cells (Tregs), their reduced response to regulation may also contribute to the unchecked effector T-cell activity in patients with MS. These concepts will be discussed below.
Collapse
Affiliation(s)
- Belinda J Kaskow
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Clare Baecher-Allan
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
40
|
Planas R, Metz I, Martin R, Sospedra M. Detailed Characterization of T Cell Receptor Repertoires in Multiple Sclerosis Brain Lesions. Front Immunol 2018; 9:509. [PMID: 29616027 PMCID: PMC5867461 DOI: 10.3389/fimmu.2018.00509] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
The antigen-specific activation of pathogenic T cells is considered essential in the initiation and maintenance of multiple sclerosis (MS). The site of activation, the differential involvement of CD4+, and CD8+ T cells, their functional phenotype, and specificity, are important aspects to understand MS pathogenesis. The analysis of clonal expansions of brain-infiltrating T cells may reveal local antigen-driven activation or specific brain homing and allow the identification of putatively pathogenic T cells. We used high-throughput T cell receptor β-chain variable gene (TRBV) sequencing (-seq) of genomic (g)DNA, which reflects the quantity and diversity of the TRBV repertoire, to characterize three white matter demyelinating lesions with different location and inflammatory activity, and paired peripheral blood memory CD4+ and CD8+ T cell pools from a secondary progressive (SP)MS patient. Our results revealed an important sharing of clonally expanded T cells with identical TRBV sequence (clonotypes) across MS lesions independently of their proximity or inflammatory activity. Comparison with circulating T cells showed that the most frequent brain-infiltrating CD8+, but not CD4+ clonotypes were also those with highest frequency in the peripheral blood, indicating clonal expansion inside the brain or specific brain homing of CD4+ but not CD8+ T cells. Parallel TRBV-seq of complementary (c)DNA that reflects the activation status of the cells, revealed differences between lesions regarding inflammatory activity and appears to facilitate the identification of putatively pathogenic T cells in active lesions. Approaches to identify pathogenic T cells in brain lesions using TRBV-seq may benefit from focusing on lesions with high inflammatory activity and from combining gDNA and cDNA sequencing.
Collapse
Affiliation(s)
- Raquel Planas
- Neuroimmunology and MS Research (nims), Department of Neurology, University Zurich, Zürich, Switzerland
| | - Imke Metz
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Roland Martin
- Neuroimmunology and MS Research (nims), Department of Neurology, University Zurich, Zürich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research (nims), Department of Neurology, University Zurich, Zürich, Switzerland
| |
Collapse
|
41
|
Hu JM, Liang WH, Qi CH, Wang XL, Pan XL, Qi LW, Shen XH, Li JF, Xie YF, Pang LJ, Liu CX, Zhang HJ, Tao L, Li F. HLA-DQB1*03 and DRB1*07 alleles increase the risk of cervical cancer among Uighur and Han women in Xinjiang, China. Future Oncol 2018. [PMID: 29513033 DOI: 10.2217/fon-2018-0048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To explore the association between the determinant factors including HLA-DQB1*03, DRB1-*07, -*13 and high-risk HPV infection, the cervical squamous cell carcinoma (CSCC) pathogenesis among Chinese Uighur and Han population. MATERIALS & METHODS HLA alleles were genotyped by PCR sequence-specific primers. RESULTS HPV16 infection rate was significantly higher among the Uighurs and Hans with CSCC as compared with healthy controls, respectively. HLA-DQB1*03 significantly increased among Uighurs with CSCC, while HLA-DRB1*07 significantly increased among Hans with CSCC. Similar tendencies were observed for DQB1*03 with HPV16-positive Uighurs CSCC and DRB1*07 with HPV16-positive Hans CSCC. CONCLUSION This study suggests that HLA-DQB1*03 and DRB1*07 alleles may influence the immune response to HPV16 infection and increase the risk of CSCC among the Uighurs and Hans in China.
Collapse
Affiliation(s)
- Jian Ming Hu
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China.,Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Wei Hua Liang
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China
| | - Cui Hua Qi
- Department of Digestion, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Xue Li Wang
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China.,Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Xiao Lin Pan
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China
| | - Li Wen Qi
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637100, PR China
| | - Xi Hua Shen
- Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Jiang Fen Li
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China.,Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Yu Fang Xie
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China.,Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Li Juan Pang
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China
| | - Chun Xia Liu
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China
| | - Hai Jun Zhang
- Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832003, PR China
| | - Lin Tao
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China
| | - Feng Li
- Department of Pathology & Key Laboratory of Xinjiang Endemic & Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, 832003, PR China.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, PR China
| |
Collapse
|
42
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
43
|
Human leucocyte antigen (HLA) class I and II typing in Belgian multiple sclerosis patients. Acta Neurol Belg 2017; 117:61-65. [PMID: 27797002 DOI: 10.1007/s13760-016-0716-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/19/2016] [Indexed: 10/20/2022]
Abstract
This is one of the first studies to compare the frequencies of different human leucocyte antigen (HLA) class I and II alleles and haplotype HLA-DRB1*15-DQB1*06 in a cohort of 119 patients with multiple sclerosis (MS) and a cohort of 124 healthy controls in Belgium. An association with MS was found for the HLA-DRB1*15 (odds ratio [OR] 2.60 [95% confidence interval (CI) 1.51-4.50]) and HLA-DQB1*06 (OR 1.97 [95% CI 1.18-3.29]) alleles, and for haplotype DRB1*15-DQB1*06 (OR 2.63 [95% CI 1.52-4.56]). The HLA-B*07 allele also tended to be more frequent in MS patients (OR 1.46 [95% CI 0.80-2.65]) and more frequent among MS patients with than in those without the HLA-DRB1*15 allele (26/54 [48.1%] versus 6/65 [9.2%]; p value <0.0001). Other alleles were underrepresented in MS patients, such as the HLA-DRB1*07 (OR 0.39 [95% CI 0.21-0.73]) and HLA-A*02 (OR 0.56 [95% CI 0.34-0.94]), showing a protective role against the disease. The HLA-B*44 (OR 0.58 [95% CI 0.31-1.09]) and HLA-DRB1*04 (OR 0.75 [95% CI 0.42-1.34]) alleles tended to be less frequent in MS patients. Altogether, the significant results observed in this population are in line with those from other countries and confirm that propensity to MS can be due to a complex presence of various HLA class I and class II alleles.
Collapse
|
44
|
Lysandropoulos AP, Mavroudakis N, Pandolfo M, El Hafsi K, van Hecke W, Maertens A, Billiet T, Ribbens A. HLA genotype as a marker of multiple sclerosis prognosis: A pilot study. J Neurol Sci 2017; 375:348-354. [PMID: 28320165 DOI: 10.1016/j.jns.2017.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/26/2016] [Accepted: 02/07/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE The identification of a biomarker with prognostic value is an unmet need in multiple sclerosis (MS). The objective of this study was to investigate a possible association of HLA genotype with disease status and progression in MS, based on comprehensive and sensitive clinical and magnetic resonance imaging (MRI) parameters to measure disease effects. METHOD A total of 118 MS patients (79 females, 39 males) underwent HLA typing. Patient MS status was assessed at two time points in a 2-year interval, based on clinical scores (including EDSS, MSSS, T25FW, 9-HPT, SDMT, BVMT, CVLT-II) and MRI evaluations. Quantitative brain MRI values were obtained for whole brain atrophy, FLAIR lesion volume change and number of new lesions using MSmetrix. Predefined HLA patient groups were compared as of disease status and progression. Global assessment was achieved by an overall t-statistic and assessment per measurement by a Welch test and/or Mann Whitney U test. The effects of multiple covariates, including age, gender and disease duration as well as scan parameters, were also evaluated using a regression analysis. RESULTS The HLA-A*02 allele was associated with better outcomes in terms of MSSS, EDSS and new lesion count (Welch test p-value<0.05). The HLA-B*07 and HLA-B*44 alleles showed a global negative effect on disease status, although none of the measurements reached significance (p-value<0.05). Results for the HLA-DRB1*15, HLA-DQB1*06 and HLA-B*08 alleles were inconclusive. The influence of the confounding variables on the statistical analysis was limited.
Collapse
Affiliation(s)
| | - Nicolas Mavroudakis
- Department of Neurology, Hôpital Erasme, Université Libre de Bruxelles, Belgium
| | - Massimo Pandolfo
- Department of Neurology, Hôpital Erasme, Université Libre de Bruxelles, Belgium
| | - Kaoutar El Hafsi
- Department of Neurology, Hôpital Erasme, Université Libre de Bruxelles, Belgium
| | | | | | | | | |
Collapse
|
45
|
Dandekar S, Wijesuriya H, Geiger T, Hamm D, Mathern GW, Owens GC. Shared HLA Class I and II Alleles and Clonally Restricted Public and Private Brain-Infiltrating αβ T Cells in a Cohort of Rasmussen Encephalitis Surgery Patients. Front Immunol 2016; 7:608. [PMID: 28066418 PMCID: PMC5165278 DOI: 10.3389/fimmu.2016.00608] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/01/2016] [Indexed: 11/26/2022] Open
Abstract
Rasmussen encephalitis (RE) is a rare pediatric neuroinflammatory disease characterized by intractable seizures and unilateral brain atrophy. T cell infiltrates in affected brain tissue and the presence of circulating autoantibodies in some RE patients have indicated that RE may be an autoimmune disease. The strongest genetic links to autoimmunity reside in the MHC locus, therefore, we determined the human leukocyte antigen (HLA) class I and class II alleles carried by a cohort of 24 RE surgery cases by targeted in-depth genomic sequencing. Compared with a reference population the allelic frequency of three alleles, DQA1*04:01:01, DQB1*04:02:01, and HLA-C*07:02:01:01 indicated that they might confer susceptibility to the disease. It has been reported that HLA-C*07:02 is a risk factor for Graves disease. Further, eight patients in the study cohort carried HLA-A*03:01:01:01, which has been linked to susceptibility to multiple sclerosis. Four patients carried a combination of three HLA class II alleles that has been linked to type 1 diabetes (DQA1*05:01:01:01~DQB1*02:01:01~DRB1*03:01:01:01), and five patients carried a combination of HLA class II alleles that has been linked to the risk of contracting multiple sclerosis (DQA1*01:02:01:01, DQB1*06:02:01, DRB1*15:01:01:01). We also analyzed the diversity of αβ T cells in brain and blood specimens from 14 of these RE surgery cases by sequencing the third complementarity regions (CDR3s) of rearranged T cell receptor β genes. A total of 31 unique CDR3 sequences accounted for the top 5% of all CDR3 sequences in the 14 brain specimens. Thirteen of these sequences were found in sequencing data from healthy blood donors; the remaining 18 sequences were patient specific. These observations provide evidence for the clonal expansion of public and private T cells in the brain, which might be influenced by the RE patient’s HLA haplotype.
Collapse
Affiliation(s)
- Sugandha Dandekar
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles , Los Angeles, CA , USA
| | - Hemani Wijesuriya
- Pathology and Laboratory Medicine, David Geffen School of Medicine at the University of California Los Angeles , Los Angeles, CA , USA
| | - Tim Geiger
- Adaptive Biotechnologies Inc. , Seattle, WA , USA
| | - David Hamm
- Adaptive Biotechnologies Inc. , Seattle, WA , USA
| | - Gary W Mathern
- Department of Neurosurgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA; Brain Research Institute, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA; Mattel Children's Hospital, Los Angeles, CA, USA
| | - Geoffrey C Owens
- Department of Neurosurgery, David Geffen School of Medicine at the University of California Los Angeles , Los Angeles, CA , USA
| |
Collapse
|
46
|
Association of HLA-A, B, DRB1* and DQB1* alleles and haplotypes in south Indian T2DM patients. Gene 2016; 592:200-208. [DOI: 10.1016/j.gene.2016.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/02/2016] [Accepted: 08/02/2016] [Indexed: 12/17/2022]
|
47
|
Brea EJ, Oh CY, Manchado E, Budhu S, Gejman RS, Mo G, Mondello P, Han JE, Jarvis CA, Ulmert D, Xiang Q, Chang AY, Garippa RJ, Merghoub T, Wolchok JD, Rosen N, Lowe SW, Scheinberg DA. Kinase Regulation of Human MHC Class I Molecule Expression on Cancer Cells. Cancer Immunol Res 2016; 4:936-947. [PMID: 27680026 DOI: 10.1158/2326-6066.cir-16-0177] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022]
Abstract
The major histocompatibility complex I (MHC-1) presents antigenic peptides to tumor-specific CD8+ T cells. The regulation of MHC-I by kinases is largely unstudied, even though many patients with cancer are receiving therapeutic kinase inhibitors. Regulators of cell-surface HLA amounts were discovered using a pooled human kinome shRNA interference-based approach. Hits scoring highly were subsequently validated by additional RNAi and pharmacologic inhibitors. MAP2K1 (MEK), EGFR, and RET were validated as negative regulators of MHC-I expression and antigen presentation machinery in multiple cancer types, acting through an ERK output-dependent mechanism; the pathways responsible for increased MHC-I upon kinase inhibition were mapped. Activated MAPK signaling in mouse tumors in vivo suppressed components of MHC-I and the antigen presentation machinery. Pharmacologic inhibition of MAPK signaling also led to improved peptide/MHC target recognition and killing by T cells and TCR-mimic antibodies. Druggable kinases may thus serve as immediately applicable targets for modulating immunotherapy for many diseases. Cancer Immunol Res; 4(11); 936-47. ©2016 AACR.
Collapse
Affiliation(s)
- Elliott J Brea
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Claire Y Oh
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Eusebio Manchado
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Sadna Budhu
- Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Ron S Gejman
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - George Mo
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Patrizia Mondello
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - James E Han
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Casey A Jarvis
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - David Ulmert
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Qing Xiang
- RNAi Core Facility, Memorial Sloan Kettering Cancer Center New York, New York
| | - Aaron Y Chang
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Ralph J Garippa
- RNAi Core Facility, Memorial Sloan Kettering Cancer Center New York, New York
| | - Taha Merghoub
- Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Jedd D Wolchok
- Weill Cornell Medicine, New York, New York.,Immunology Program, Memorial Sloan Kettering Cancer Center New York, New York
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York.,Weill Cornell Medicine, New York, New York
| | - Scott W Lowe
- Weill Cornell Medicine, New York, New York.,Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center New York, New York.,Howard Hughes Medical Institute, New York, New York
| | - David A Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center New York, New York. .,Weill Cornell Medicine, New York, New York
| |
Collapse
|
48
|
Mathias A, Perriard G, Canales M, Soneson C, Delorenzi M, Schluep M, Du Pasquier RA. Increased ex vivo antigen presentation profile of B cells in multiple sclerosis. Mult Scler 2016; 23:802-809. [PMID: 27503907 DOI: 10.1177/1352458516664210] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is thought to be T cell mediated but the mechanisms eliciting such a dysregulated adaptative immune response remain enigmatic. OBJECTIVE To examine the activation profile of antigen-presenting cells (APCs) in MS. METHODS A total of 98 study subjects were enrolled including patients suffering from relapsing-remitting, secondary- and primary-progressive (PP) MS, other inflammatory neurological diseases, and healthy controls. Blood monocytes and B cells were stimulated using specific ligands of toll-like receptors (TLRs) or inflammasomes or Epstein-Barr virus (EBV) particles. Their activation profile was determined before or after stimulation by flow cytometry (CD40, CD80, CD83, CD86, and human leukocyte antigen-antigen D related (HLA-DR)) and Luminex assay, measuring the concentration of eight cytokines in culture supernatants. Differences among groups were assessed in a linear model framework. RESULTS We demonstrate that relapsing MS patients exhibit an increased expression of HLA-DR and CD40 ex vivo, mostly at the surface of B cells. Specific stimulations of TLR or inflammasomes enhance the expression of components of the immunological synapse and the cytokine secretion but without differences between categories of study subjects. CONCLUSION These data suggest that the activation profile of B cells is increased in MS. However, the perception of the danger signal by B lymphocytes and monocytes does not seem to be different in MS patients as compared to control subjects.
Collapse
Affiliation(s)
- Amandine Mathias
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Guillaume Perriard
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Mathieu Canales
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Charlotte Soneson
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mauro Delorenzi
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland/Ludwig Center for Cancer Research and Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Myriam Schluep
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Renaud A Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland/Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
49
|
da Costa DSMM, Hygino J, Ferreira TB, Kasahara TM, Barros PO, Monteiro C, Oliveira A, Tavares F, Vasconcelos CC, Alvarenga R, Bento CAM. Vitamin D modulates different IL-17-secreting T cell subsets in multiple sclerosis patients. J Neuroimmunol 2016; 299:8-18. [PMID: 27725127 DOI: 10.1016/j.jneuroim.2016.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
Vitamin D deficiency is an environmental risk factor for MS, a Th17 cell-mediated autoimmune disease that results in demyelination in the CNS. Therefore, we aimed to evaluate the ability of in vitro 1,25(OH)2D in modulating different Th17 cell subsets in MS patients in remission phase. In the present study, the production of Th17-related cytokines (IL-1β, IL-6, IL-17, IL-22), as well as GM-CSF, was significantly higher in cell cultures from MS patients than in healthy subjects (HS). The 1,25(OH)2D reduced all pro-inflammatory cytokines essayed, mainly those released from HS cell cultures. The proportion of both IL-17+IFN-γ+ (CD4+ and CD8+) T cells and IL-17+IFN-γ-CD8+ T cells was positively related with neurological disorders, determined by EDSS score. The addition of 1,25(OH)2D reduced not only these pathogenic T cell subsets but elevated the percentage of IL-10-secreting conventional (FoxP3+CD25+CD127-CD4+) and non-conventional (IL-17+) regulatory-like T cells. Taken together, the results indicate that the active form of vitamin D should benefit MS patients by attenuating the percentage of pathogenic T cells. This effect could be direct and/or indirect, by enhancing classical and non-classical regulatory T cells.
Collapse
Affiliation(s)
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Thais B Ferreira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Priscila O Barros
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Aleida Oliveira
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | - Felipe Tavares
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil
| | | | - Regina Alvarenga
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Post-graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Brazil; Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
50
|
Reiss CS. Virus-Induced Demyelination: The Case for Virus(es) in Multiple Sclerosis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7122906 DOI: 10.1007/978-3-319-33189-8_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple Sclerosis (MS) is the most common demyelinating disease of man with over 400,000 cases in the United States and over 2.5 million cases worldwide. There are over 64,000 citations in Pubmed dating back as far as 1887. Much has been learned over the past 129 years with a recent burst in therapeutic options (mostly anti-inflammatory) with newer medications in development that are neuroprotective and/or neuroreparative. However, with all these advancements the cause of MS remains elusive. There is a clear interplay of genetic, immunologic, and environmental factors that influences both the development and progression of this disorder. This chapter will give a brief overview of the history and pathogenesis of MS with attention to how host immune responses in genetically susceptible individuals contribute to the MS disease process. In addition, we will explore the role of infectious agents in MS as potential “triggers” of disease. Models of virus-induced demyelination will be discussed, with an emphasis on the recent interest in human herpesviruses and the role they may play in MS disease pathogenesis. Although we remain circumspect as to the role of any microbial pathogen in MS, we suggest that only through well-controlled serological, cellular immune, molecular, and animal studies we will be able to identify candidate agents. Ultimately, clinical interventional trials that either target a specific pathogen or class of pathogens will be required to make definitive links between the suspected agent and MS.
Collapse
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|