1
|
Li J, Mao N, Wang Y, Deng S, Chen K. Novel insights into the ROCK-JAK-STAT signaling pathway in upper respiratory tract infections and neurodegenerative diseases. Mol Ther 2025; 33:32-50. [PMID: 39511889 DOI: 10.1016/j.ymthe.2024.11.011] [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: 08/12/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Acute upper respiratory tract infections are a major public health issue, with uncontrolled inflammation triggered by upper respiratory viruses being a significant cause of patient deterioration or death. This study focuses on the Janus kinase-signal transducer and activator of transcription Rho-associated coiled-coil containing protein kinase (JAK-STAT-ROCK) signaling pathway, providing an in-depth analysis of the interplay between uncontrolled inflammation after upper respiratory tract infections and the development of neurodegenerative diseases. It offers a conceptual framework for understanding the lung-brain-related immune responses and potential interactions. The relationship between the ROCK-JAK-STAT signaling pathway and inflammatory immunity is a complex and multi-layered research area and exploring potential common targets could open new avenues for the prevention and treatment of related inflammation.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Naihui Mao
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China.
| |
Collapse
|
2
|
Vida H, Sahar M, Nikdouz A, Arezoo H. Chemokines in neurodegenerative diseases. Immunol Cell Biol 2024. [PMID: 39723647 DOI: 10.1111/imcb.12843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/09/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Neurodegeneration and neuroinflammation disorders are mainly the result of the deposition of various proteins, such as α-synuclein, amyloid-β and prions, which lead to the initiation and activation of inflammatory responses. Different chemokines are involved in the infiltration and movement of inflammatory leukocytes into the central nervous system (CNS) that express chemokine receptors. Dysregulation of several members of chemokines has been shown in the CNS, cerebrospinal fluid and peripheral blood of patients who have neurodegenerative disorders. Upon infiltration of various cells, they produce many inflammatory mediators such as cytokines. Besides them, some CNS-resident cells, such as neurons and astrocytes, are also involved in the pathogenesis of neurodegeneration by producing chemokines. In this review, we summarize the role of chemokines and their related receptors in the pathogenesis of neurodegeneration and neuroinflammation disorders, including multiple sclerosis, Parkinson's disease and Alzheimer's disease. Therapeutic strategies targeting chemokines or their related receptors are also discussed in this article.
Collapse
Affiliation(s)
- Hashemi Vida
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mehranfar Sahar
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Urmia University of Medical Sciences, Urmia, Iran
| | - Amin Nikdouz
- Department of Translational Medicine, Universita degli Studi del Piemonte Orientale Amedeo Avogadro, Vercelli, Italy
| | - Hosseini Arezoo
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
3
|
Proschinger S, Belen S, Adammek F, Schlagheck ML, Rademacher A, Schenk A, Warnke C, Bloch W, Zimmer P. Sportizumab - Multimodal progressive exercise over 10 weeks decreases Th17 frequency and CD49d expression on CD8 + T cells in relapsing-remitting multiple sclerosis: A randomized controlled trial. Brain Behav Immun 2024; 124:397-408. [PMID: 39675643 DOI: 10.1016/j.bbi.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) represents a neuroinflammatory autoimmune disease characterized by the predominance of circulating T cell subsets with proinflammatory characteristics and increased central nervous system (CNS)-homing potential. Substantial evidence confirms various beneficial effects of chronic exercise interventions in MS, but it is unknown how long-term multi-modal intense exercise affects MS-associated lymphocytes that are commonly targeted by medication in persons with relapsing remitting MS (pwRRMS). METHODS A total of 45 participants with defined RRMS were randomized to either the exercise (n = 22) or passive waitlist-control group (n = 23). A 10-week intervention consisting of progressive resistance and strength-endurance exercises was applied (3x/week à 60 min). Blood was drawn before (T1) and after (T2) the intervention period. Flow cytometry was used for phenotyping lymphocyte subsets. RESULTS Relative protein expression of CD49d within CD8+ T cells, quantified via mean fluorescence intensity (MFI), is significantly associated with the Expanded Disability Status Scale (p = 0.007, r = 0.440), decreased in the exercise group (p = 0.001) only, and was significantly lower in the exercise compared to the control group at T2 (p < 0.001). T helper (Th) 17 cell frequency decreased only in the exercise group (p < 0.001). CD8+CD20+ T cell frequency was significantly lower in the exercise compared to the control group at T2 (p = 0.003), without showing significant time effects. CONCLUSION The 10-week multimodal exercise intervention mainly affected circulating T cells harboring a pathophysiological phenotype in MS. The findings of a decreased frequency of pathogenic Th17 cells and the reduced CNS-homing potential of CD8+ T cells, indicated by reduced CD49d MFI, substantiate the positive effects of exercise on cellular biomarkers involved in disease activity and progression in MS. To confirm exercise-mediated beneficial effects on both disease domains, clinical endpoints (i.e., relapse rate, lesion formation, EDSS score) should be assessed together with these cellular and molecular markers in studies with a larger sample size and a duration of six to twelve months or longer.
Collapse
Affiliation(s)
- Sebastian Proschinger
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Sergen Belen
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany; Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Frederike Adammek
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Marit Lea Schlagheck
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | | | - Alexander Schenk
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Clemens Warnke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany.
| |
Collapse
|
4
|
Lei X, Xiao R, Chen Z, Ren J, Zhao W, Tang W, Wen K, Zhu Y, Li X, Ouyang S, Xu A, Hu Y, Bi E. Augmenting antitumor efficacy of Th17-derived Th1 cells through IFN-γ-induced type I interferon response network via IRF7. Proc Natl Acad Sci U S A 2024; 121:e2412120121. [PMID: 39541355 PMCID: PMC11588128 DOI: 10.1073/pnas.2412120121] [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: 06/19/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The importance of CD4+ T cells in cancer immunotherapy has gained increasing recognition. Particularly, a specific subset of CD4+ T cells coexpressing the T helper type 1 (Th1) and Th17 markers has demonstrated remarkable antitumor potential. However, the underlying mechanisms governing the differentiation of these cells and their subsequent antitumor responses remain incompletely understood. Single-cell RNA sequencing (scRNA-seq) data reanalysis demonstrated the presence of Th171 cells within tumors. Subsequent trajectory analysis found that these Th171 cells are initially primed under Th17 conditions and then converted into IFN-γ-producing cells. Following the in vivo differentiation trajectory of Th171 cells, we successfully established in vitro Th171 cell culture. Transcriptomic profiling has unveiled a substantial resemblance between in vitro-generated Th171 cells and their tumor-infiltrating counterparts. Th171 cells exhibit more potent antitumor responses than Th1 or Th17 cells. Additionally, Th171chimeric antigen receptor T (CAR-T) cells eradicate solid tumors more efficiently. Importantly, Th171 cells display an early exhaustion phenotype while retaining stemness. Mechanistically, Th171 cells migrate faster and accumulate more in tumors in an extracellular matrix protein 1 (ECM1)-dependent manner. Furthermore, we show that IFN-γ up-regulated IRF7 to promote the type I interferon response network and ECM1 expression but decreased the exhaustion status in Th171 cells. Taken together, our findings position Th171 cells as a great candidate for improving targeted immunotherapies in solid malignancies.
Collapse
Affiliation(s)
- Xiaoyi Lei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Ruipei Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Zhe Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenli Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wenting Tang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Kang Wen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yihan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xinru Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan523808, China
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| | - Yu Hu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou510515, China
| | - Enguang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong510515, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou510280, China
| |
Collapse
|
5
|
Räuber S, Schulte-Mecklenbeck A, Willison A, Hagler R, Jonas M, Pul D, Masanneck L, Schroeter CB, Golombeck KS, Lichtenberg S, Strippel C, Gallus M, Dik A, Kerkhoff R, Barman S, Weber KJ, Kovac S, Korsen M, Pawlitzki M, Goebels N, Ruck T, Gross CC, Paulus W, Reifenberger G, Hanke M, Grauer O, Rapp M, Sabel M, Wiendl H, Meuth SG, Melzer N. Flow cytometry identifies changes in peripheral and intrathecal lymphocyte patterns in CNS autoimmune disorders and primary CNS malignancies. J Neuroinflammation 2024; 21:286. [PMID: 39497174 PMCID: PMC11536547 DOI: 10.1186/s12974-024-03269-3] [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: 09/15/2024] [Accepted: 10/20/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Immune dysregulation is a hallmark of autoimmune diseases of the central nervous system (CNS), characterized by an excessive immune response, and primary CNS tumors (pCNS-tumors) showing a highly immunosuppressive parenchymal microenvironment. METHODS Aiming to provide novel insights into the pathogenesis of CNS autoimmunity and cerebral tumor immunity, we analyzed the peripheral blood (PB) and cerebrospinal fluid (CSF) of 81 autoimmune limbic encephalitis (ALE), 148 relapsing-remitting multiple sclerosis (RRMS), 33 IDH-wildtype glioma, 9 primary diffuse large B cell lymphoma of the CNS (CNS-DLBCL), and 110 controls by flow cytometry (FC). Additionally, an in-depth immunophenotyping of the PB from an independent cohort of 20 RRMS and 18 IDH-wildtype glioblastoma patients compared to 19 controls was performed by FC combined with unsupervised computational approaches. RESULTS We identified alterations in peripheral and intrathecal adaptive immunity, mainly affecting the T cell (Tc) but also the B cell (Bc) compartment in ALE, RRMS, and pCNS-tumors compared to controls. ALE, RRMS, and pCNS-tumors featured higher expression of the T cell activation marker HLA-DR, which was even more pronounced in pCNS-tumors than in ALE or RRMS. Glioblastoma patients showed signs of T cell exhaustion that were not visible in RRMS patients. In-depth characterization of the PB revealed differences mainly in the T effector and memory compartment between RRMS and glioblastoma patients and similar alterations in the Bc compartment, including atypical Bc, CD19+CD20- double negative Bc, and plasma cells. PB and CSF mFC together with CSF routine parameters could reliably differentiate ALE and RRMS from pCNS-tumors facilitating early diagnosis and treatment. CONCLUSIONS ALE, RRMS, and pCNS-tumors show distinct but partially overlapping changes mainly in HLA-DR+ Tc, memory Tc, exhausted Tc, and Bc subsets providing insights into disease pathogenesis. Moreover, mFC shows diagnostic potential facilitating early diagnosis and treatment.
Collapse
Affiliation(s)
- Saskia Räuber
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | | | - Alice Willison
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Ramona Hagler
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Marius Jonas
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Duygu Pul
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Lars Masanneck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Kristin S Golombeck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Stefanie Lichtenberg
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Christine Strippel
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Marco Gallus
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Andre Dik
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Ruth Kerkhoff
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Sumanta Barman
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Katharina J Weber
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurological Institute (Edinger Institute), University Hospital, Goethe University, Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Melanie Korsen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Werner Paulus
- Institute of Neuropathology, University of Münster, Münster, Germany
| | - Guido Reifenberger
- Institute of Neuropathology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Hanke
- Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Center Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Grauer
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Marion Rapp
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Sabel
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Nico Melzer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany.
| |
Collapse
|
6
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
7
|
Abbasi Kasbi N, Ghane Ezabadi S, Kohandel K, Khodaie F, Sahraian AH, Nikkhah Bahrami S, Mohammadi M, Almasi-Hashiani A, Eskandarieh S, Sahraian MA. Lifetime exposure to smoking and substance abuse may be associated with late-onset multiple sclerosis: a population-based case-control study. BMC Neurol 2024; 24:327. [PMID: 39243006 PMCID: PMC11378646 DOI: 10.1186/s12883-024-03815-9] [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: 01/21/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Late-onset multiple sclerosis (LOMS), defined as the development of MS after the age of 50, has shown a substantial surge in incidence rates and is associated with more rapid progression of disability. Besides, studies have linked tobacco smoking to a higher chance of MS progression. However, the role of smoking on the risk of developing LOMS remains unclear. This study aims to evaluate the possible association between lifetime exposure to cigarette and waterpipe smoking, drug abuse, and alcohol consumption and the risk of LOMS. METHODS This population-based case-control study involved LOMS cases and healthy sex and age-matched controls from the general population in Tehran, Iran. The primary data for confirmed LOMS cases were obtained from the nationwide MS registry of Iran (NMSRI), while supplementary data were collected through telephone and on-site interviews. Predesigned questionnaire for multinational case-control studies of MS environmental risk factors was used to evaluate the LOMS risk factors. The study employed Likelihood ratio chi-square test to compare qualitative variables between the two groups and utilized two independent sample t-test to compare quantitative data. Adjusted odds ratio (AOR) for age along with 95% confidence intervals (CI) were calculated using matched logistic regression analysis in SPSS 23. RESULTS Totally, 83 LOMS cases and 207 controls were included in the analysis. The female to male ratio in the cases was 1.5: 1. The mean ± SD age of 83 cases and 207 controls was 61.14 ± 5.38) and 61.51 ± 7.67 years, respectively. The mean ± SD expanded disability status scale (EDSS) score was 3.68 ± 2.1. Although the results of waterpipe exposure had no significant effect on LOMS development (P-value: 0.066), ever cigarette-smoked participants had a significantly higher risk of developing LOMS than those who never smoked (AOR: 2.57, 95% CI: 1.44-4.60). Furthermore, people with a history of smoking for more than 20 years had 3.45 times the odds of developing MS than non-smokers. Drug and alcohol abuse were both associated with LOMS in our study; of which opioids (AOR: 5.67, 95% CI: 2.05-15.7), wine (AOR: 3.30, 95% CI: 1.41-7.71), and beer (AOR: 3.12, 95% CI: 1.45-6.69) were found to pose the greatest risk of LOMS, respectively. CONCLUSION For the first time, we identified smoking, drug, and alcohol use as potential risk factors for LOMS development. According to the global increase in cigarette smoking and alcohol use, these findings highlight the importance of conducting interventional approaches for prevention.
Collapse
Affiliation(s)
- Naghmeh Abbasi Kasbi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Sajjad Ghane Ezabadi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Kosar Kohandel
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Faezeh Khodaie
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Amir Hossein Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Sahar Nikkhah Bahrami
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
- Student Research Committee, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Mohammadi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Sharareh Eskandarieh
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran.
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Hasan Abad Sq., Tehran, Iran.
| |
Collapse
|
8
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
9
|
Hu H, Li H, Li R, Liu P, Liu H. Re-establishing immune tolerance in multiple sclerosis: focusing on novel mechanisms of mesenchymal stem cell regulation of Th17/Treg balance. J Transl Med 2024; 22:663. [PMID: 39010157 PMCID: PMC11251255 DOI: 10.1186/s12967-024-05450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
The T-helper 17 (Th17) cell and regulatory T cell (Treg) axis plays a crucial role in the development of multiple sclerosis (MS), which is regarded as an immune imbalance between pro-inflammatory cytokines and the maintenance of immune tolerance. Mesenchymal stem cell (MSC)-mediated therapies have received increasing attention in MS research. In MS and its animal model experimental autoimmune encephalomyelitis, MSC injection was shown to alter the differentiation of CD4+T cells. This alteration occurred by inducing anergy and reduction in the number of Th17 cells, stimulating the polarization of antigen-specific Treg to reverse the imbalance of the Th17/Treg axis, reducing the inflammatory cascade response and demyelination, and restoring an overall state of immune tolerance. In this review, we summarize the mechanisms by which MSCs regulate the balance between Th17 cells and Tregs, including extracellular vesicles, mitochondrial transfer, metabolic reprogramming, and autophagy. We aimed to identify new targets for MS treatment using cellular therapy by analyzing MSC-mediated Th17-to-Treg polarization.
Collapse
Affiliation(s)
- Huiru Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Hui Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ruoyu Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Peidong Liu
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Hongbo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
10
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
11
|
Zrzavy T, Rieder K, Wuketich V, Thalhammer R, Haslacher H, Altmann P, Kornek B, Krajnc N, Monschein T, Schmied C, Zebenholzer K, Zulehner G, Berger T, Rommer P, Leutmezer F, Bsteh G. Immunophenotyping in routine clinical practice for predicting treatment response and adverse events in patients with MS. Front Neurol 2024; 15:1388941. [PMID: 38689880 PMCID: PMC11058637 DOI: 10.3389/fneur.2024.1388941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Background Recent studies proposed cellular immunoprofiling as a surrogate for predicting treatment response and/or stratifying the occurrence of adverse events (AEs) in persons with multiple sclerosis (pwMS). However, applicability in real-world circumstances is not sufficiently addressed. Objective We aimed to explore whether standard routine clinical leukocyte phenotyping before treatment initiation could help stratify patients according to treatment response or AEs in a real-world MS cohort. Methods In this retrospective study, 150 pwMS were included, who had been newly initiated on a disease-modifying drug (DMD) and had been assessed for standard immunophenotyping before DMD initiation (baseline) and at least once during the following year. Multivariate models were used to assess an association of immune subsets and the association between immune cell profiles regarding treatment response and AEs. Results We found that the composition of T cell subsets was associated with relapse activity, as an increased proportion of CD8+ lymphocytes at baseline indicated a higher likelihood of subsequent relapse (about 9% per 1% increase in CD8+ proportion of all CD3+ cells). This was particularly driven by patients receiving anti-CD20 therapy, where also EDSS worsening was associated with a higher number of CD8+ cells at baseline (3% increase per 10 cells). In the overall cohort, an increase in the proportion of NK cells was associated with a higher risk of EDSS worsening (5% per 1% increase). Occurrence of AEs was associated with a higher percentage of T cells and a lower number of percentual NKT cells at baseline. Conclusion Immune cell profiles are associated with treatment response and the occurrence of AEs in pwMS. Hence, immunophenotyping may serve as a valuable biomarker to enable individually tailored treatment strategies in pwMS.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Kerstin Rieder
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Viktoria Wuketich
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Patrick Altmann
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Barbara Kornek
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Tobias Monschein
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Christiane Schmied
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Karin Zebenholzer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Gudrun Zulehner
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Paulus Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Fritz Leutmezer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Peng Y, Zhang X, Tang Y, He S, Rao G, Chen Q, Xue Y, Jin H, Liu S, Zhou Z, Xiang Y. Role of autoreactive Tc17 cells in the pathogenesis of experimental autoimmune encephalomyelitis. NEUROPROTECTION 2024; 2:49-59. [DOI: 10.1002/nep3.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 07/04/2024]
Abstract
AbstractBackgroundThe pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE—an animal model of MS) is primarily mediated by T cells. However, recent studies have only focused on interleukin (IL)‐17‐secreting CD4+ T‐helper cells, also known as Th17 cells. This study aimed to compare Th17 cells and IL‐17‐secreting CD8+ T‐cytotoxic cells (Tc17) in the context of MS/EAE.MethodsFemale C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein peptides 35–55 (MOG35–55), pertussis toxin, and complete Freund's adjuvant to establish the EAE animal model. T cells were isolated from the spleen (12–14 days postimmunization). CD4+ and CD8+ T cells were purified using isolation kit and then differentiated into Th17 and Tc17, respectively, using MOG35–55 and IL‐23. The secretion levels of interferon‐γ (IFN‐γ) and IL‐17 were measured via enzyme‐linked immunosorbent assay using cultured CD4+ and CD8+ T cell supernatants. The pathogenicity of Tc17 and Th17 cells was assessed through adoptive transfer (tEAE), with the clinical course assessed using an EAE score (0–5). Hematoxylin and eosin as well as Luxol fast blue staining were used to examine the spinal cord. Purified CD8+ CD3+ and CD4+ CD3+ cells differentiated into Tc17 and Th17 cells, respectively, were stimulated with MOG35–55 peptide for proliferation assays.ResultsThe results showed that Tc17 cells (15,951 ± 1985 vs. 55,709 ± 4196 cpm; p < 0.050) exhibited a weaker response to highest dose (20 μg/mL) MOG35–55 than Th17 cells. However, this response was not dependent on Th17 cells. After the 48 h stimulation, at the highest dose (20 μg/mL) of MOG35–55. Tc17 cells secreted lower levels of IFN‐γ (280.00 ± 15.00 vs. 556.67 ± 15.28 pg/mL, p < 0.050) and IL‐17 (102.67 ± 5.86 pg/mL vs. 288.33 ± 12.58 pg/mL; p < 0.050) than Th17 cells. Similar patterns were observed for IFN‐γ secretion at 96 and 144 h. Furthermore, Tc17 cell‐induced tEAE mice exhibited similar EAE scores to Th17 cell‐induced tEAE mice and also showed similar inflammation and demyelination.ConclusionThe degree of pathogenicity of Tc17 cells in EAE is lower than that of Th17 cells. Future investigation on different immune cells and EAE models is warranted to determine the mechanisms underlying MS.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Xiuli Zhang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yandan Tang
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shunqing He
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Guilan Rao
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Quan Chen
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Yahui Xue
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Hong Jin
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shu Liu
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Ziyang Zhou
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yun Xiang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| |
Collapse
|
13
|
Uriol-Rivera MG, Obrador-Mulet A, Juliá MR, Daza-Cajigal V, Delgado-Sanchez O, Garcia Alvarez A, Gomez-Lobon A, Carrillo-Garcia P, Saus-Sarrias C, Gómez-Cobo C, Ramis-Cabrer D, Gasco Company J, Molina-Infante J. Sequential administration of paricalcitol followed by IL-17 blockade for progressive refractory IgA nephropathy patients. Sci Rep 2024; 14:4866. [PMID: 38418932 PMCID: PMC10902332 DOI: 10.1038/s41598-024-55425-7] [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: 10/03/2023] [Accepted: 02/23/2024] [Indexed: 03/02/2024] Open
Abstract
There is no established treatment for progressive IgA nephropathy refractory to steroids and immunosuppressant drugs (r-IgAN). Interleukin 17 (IL-17) blockade has garnered interest in immune-mediated diseases involving the gut-kidney axis. However, single IL-17A inhibition induced paradoxical effects in patients with Crohn's disease and some cases of de novo glomerulonephritis, possibly due to the complete Th1 cell response, along with the concomitant downregulation of regulatory T cells (Tregs). Seven r-IgAN patients were treated with at least six months of oral paricalcitol, followed by the addition of subcutaneous anti-IL-17A (secukinumab). After a mean follow-up of 28 months, proteinuria decreased by 71% (95% CI: 56-87), P < 0.001. One patient started dialysis, while the annual eGFR decline in the remaining patients [mean (95% CI)] was reduced by 4.9 mL/min/1.73 m2 (95% CI: 0.1-9.7), P = 0.046. Circulating Th1, Th17, and Treg cells remained stable, but Th2 cells decreased, modifying the Th1/Th2 ratio. Intriguingly, accumulation of circulating Th17.1 cells was observed. This novel sequential therapy appears to optimize renal advantages in patients with r-IgAN and elicit alterations in potentially pathogenic T helper cells.
Collapse
Affiliation(s)
- Miguel G Uriol-Rivera
- Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain.
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain.
| | - Aina Obrador-Mulet
- Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Maria Rosa Juliá
- Immunology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Vanessa Daza-Cajigal
- Immunology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Olga Delgado-Sanchez
- Pharmacy Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Angel Garcia Alvarez
- Pharmacy Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Ana Gomez-Lobon
- Pharmacy Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Paula Carrillo-Garcia
- Pathology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Carlos Saus-Sarrias
- Pathology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
| | - Cristina Gómez-Cobo
- Laboratory Medicine Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Daniel Ramis-Cabrer
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Joan Gasco Company
- Nephrology Department, Hospital Universitario Son Espases, Palma de Mallorca, Balearic Islands, Spain
- Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | | |
Collapse
|
14
|
Holm Hansen R, von Essen MR, Reith Mahler M, Cobanovic S, Sellebjerg F. Sustained effects on immune cell subsets and autoreactivity in multiple sclerosis patients treated with oral cladribine. Front Immunol 2024; 15:1327672. [PMID: 38433828 PMCID: PMC10904620 DOI: 10.3389/fimmu.2024.1327672] [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: 10/25/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Cladribine tablet therapy is an efficacious treatment for multiple sclerosis (MS). Recently, we showed that one year after the initiation of cladribine treatment, T and B cell crosstalk was impaired, reducing potentially pathogenic effector functions along with a specific reduction of autoreactivity to RAS guanyl releasing protein 2 (RASGRP2). In the present study we conducted a longitudinal analysis of the effect of cladribine treatment in patients with RRMS, focusing on the extent to which the effects observed on T and B cell subsets and autoreactivity after one year of treatment are maintained, modulated, or amplified during the second year of treatment. Methods In this case-control exploratory study, frequencies and absolute counts of peripheral T and B cell subsets and B cell cytokine production from untreated patients with relapsing-remitting MS (RRMS) and patients treated with cladribine for 52 (W52), 60 (W60), 72 (W72) and 96 (W96) weeks, were measured using flow cytometry. Autoreactivity was assessed using a FluoroSpot assay. Results We found a substantial reduction in circulating memory B cells and proinflammatory B cell responses. Furthermore, we observed reduced T cell responses to autoantigens possibly presented by B cells (RASGRP2 and a-B crystallin (CRYAB)) at W52 and W96 and a further reduction in responses to the myelin antigens myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) after 96 weeks. Conclusion We conclude that the effects of cladribine observed after year one are maintained and, for some effects, even increased two years after the initiation of a full course of treatment with cladribine tablets.
Collapse
Affiliation(s)
- Rikke Holm Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Marina Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Mie Reith Mahler
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Stefan Cobanovic
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Raghib MF, Bao F, Elkhooly M, Bernitsas E. Choroid plexus volume as a marker of retinal atrophy in relapsing remitting multiple sclerosis. J Neurol Sci 2024; 457:122884. [PMID: 38237367 DOI: 10.1016/j.jns.2024.122884] [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: 05/30/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To evaluate choroid plexus (CP) volume as a biomarker for predicting clinical disability and retinal layer atrophy in relapsing remitting multiple sclerosis (RRMS). METHODS Ninety-five RRMS patients and 26 healthy controls (HCs) underwent 3 T whole brain MRI, expanded disability status scale (EDSS) and optical coherence tomography (OCT). Fully automated intra-retinal segmentation was performed to obtain the volumes of the retinal nerve fiber layer (RNFL), combined ganglion cell layer -inner plexiform layer (GCIPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), retinal pigment epithelium (RPE), total macular volume (TMV) and papillomacular bundle (PMB). Automated segmentation of the CP within the lateral ventricles was performed and the choroid plexus volume (CPV) was normalized by total intracranial volume (TIV). Linear regression analysis and generalized estimating equation (GEE) models were applied to evaluate relationships between nCPV and EDSS, T2 lesion volume, disease duration, and retinal layer volumes, followed by Bonferroni correction analysis for multiple comparisons. RESULTS RRMS patients had larger tChPV compared to HCs (p < 0.001). After Bonferroni correction, there was a significant positive correlation between tChPV and EDSS (r2 = 0.25, p = 0.0002), disease duration (r2 = 0.30, p = 0.01), and T2 lesion volume (r2 = 0.39, p = 0.0000). A robust negative correlation was found between tChPV and RNFL (p < 0.001), GCIPL (p = 0.003), TMV (p = 0.0185), PMB (p < 0.0001), G (p = 0.04), T(p = 0.0001). CONCLUSIONS Our findings support the association of tChPV with disability and altered retinal integrity in RRMS.
Collapse
Affiliation(s)
- Muhammad F Raghib
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Fen Bao
- Department of Neurology, Wayne State University School of Medicine, United States of America
| | - Mahmoud Elkhooly
- Department of Neurology, Wayne State University School of Medicine, United States of America; Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, United States of America; Department of Neurology and Psychiatry, Minia University, Minia, Egypt
| | - Evanthia Bernitsas
- Department of Neurology, Wayne State University School of Medicine, United States of America; Detroit Medical Center, Detroit, MI, United States of America.
| |
Collapse
|
16
|
Hoeks C, Puijfelik FV, Koetzier SC, Rip J, Corsten CEA, Wierenga-Wolf AF, Melief MJ, Stinissen P, Smolders J, Hellings N, Broux B, van Luijn MM. Differential Runx3, Eomes, and T-bet expression subdivides MS-associated CD4 + T cells with brain-homing capacity. Eur J Immunol 2024; 54:e2350544. [PMID: 38009648 DOI: 10.1002/eji.202350544] [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: 04/28/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Multiple sclerosis (MS) is a common and devastating chronic inflammatory disease of the CNS. CD4+ T cells are assumed to be the first to cross the blood-central nervous system (CNS) barrier and trigger local inflammation. Here, we explored how pathogenicity-associated effector programs define CD4+ T cell subsets with brain-homing ability in MS. Runx3- and Eomes-, but not T-bet-expressing CD4+ memory cells were diminished in the blood of MS patients. This decline reversed following natalizumab treatment and was supported by a Runx3+ Eomes+ T-bet- enrichment in cerebrospinal fluid samples of treatment-naïve MS patients. This transcription factor profile was associated with high granzyme K (GZMK) and CCR5 levels and was most prominent in Th17.1 cells (CCR6+ CXCR3+ CCR4-/dim ). Previously published CD28- CD4 T cells were characterized by a Runx3+ Eomes- T-bet+ phenotype that coincided with intermediate CCR5 and a higher granzyme B (GZMB) and perforin expression, indicating the presence of two separate subsets. Under steady-state conditions, granzyme Khigh Th17.1 cells spontaneously passed the blood-brain barrier in vitro. This was only found for other subsets including CD28- cells when using inflamed barriers. Altogether, CD4+ T cells contain small fractions with separate pathogenic features, of which Th17.1 seems to breach the blood-brain barrier as a possible early event in MS.
Collapse
Affiliation(s)
- Cindy Hoeks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Fabiënne van Puijfelik
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Steven C Koetzier
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cato E A Corsten
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Niels Hellings
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Bieke Broux
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
17
|
van Puijfelik F, Blok KM, Klein Kranenbarg RAM, Rip J, de Beukelaar J, Wierenga-Wolf AF, Wokke B, van Luijn MM, Smolders J. Ocrelizumab associates with reduced cerebrospinal fluid B and CD20 dim CD4 + T cells in primary progressive multiple sclerosis. Brain Commun 2024; 6:fcae021. [PMID: 38385000 PMCID: PMC10881107 DOI: 10.1093/braincomms/fcae021] [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: 09/25/2023] [Revised: 12/01/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
The anti-CD20 monoclonal antibody ocrelizumab reduces disability progression in primary progressive multiple sclerosis. CD20 is a prototypical B-cell marker; however, subpopulations of CD4+ and CD8+ T cells in peripheral blood and cerebrospinal fluid also express low levels of CD20 (CD20dim). Therefore, direct targeting and depletion of these CD20dim T-cell subpopulations may contribute to the therapeutic effect of ocrelizumab. The aim of this observational cohort study was to compare CD20+ B-cell and CD20dim T-cell distributions between peripheral blood and cerebrospinal fluid of ocrelizumab-treated or ocrelizumab-untreated people with primary progressive multiple sclerosis. Ocrelizumab treatment was associated with depletion of circulating B cells and CD20dim CD4+ and CD20dim CD8+ T cells (P < 0.0001, P = 0.0016 and P = 0.0008, respectively) but, in cerebrospinal fluid, only with lower proportions of B cells and CD20dim memory CD4+ T cells (P < 0.0001 and P = 0.0043, respectively). The proportional prevalence of cerebrospinal fluid CD20dim memory CD8+ T cells was not significantly reduced (P = 0.1333). Only in cerebrospinal fluid, the proportions of CD20dim cells within CD4+ and not CD8+ T cells positive for CCR5, CCR6 and CXCR3 were reduced in ocrelizumab-treated participants. The proportion of CD20dim CD4+ T cells and abundance of CD4+ relative to CD8+ T cells in cerebrospinal fluid correlated positively with age (R = 0.6799, P = 0.0150) and Age-Related Multiple Sclerosis Severity score (R = 0.8087, P = 0.0014), respectively. We conclude that, in contrast to cerebrospinal fluid CD20dim CD8+ T cells, B cells and CD20dim CD4+ T cells are reduced in cerebrospinal fluid of people with primary progressive multiple sclerosis with an ocrelizumab-associated depletion of circulating B cells and CD20dim T cells. Therefore, these cells are likely to contribute to the therapeutic effects of ocrelizumab in people with primary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Fabiënne van Puijfelik
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Katelijn M Blok
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Romy A M Klein Kranenbarg
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Janet de Beukelaar
- Department of Neurology, Albert Schweitzer Hospital, 3318 AT, Dordrecht, The Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Beatrijs Wokke
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 CN, Rotterdam, The Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Lee CY, Chan KH. Personalized Use of Disease-Modifying Therapies in Multiple Sclerosis. Pharmaceutics 2024; 16:120. [PMID: 38258130 PMCID: PMC10820407 DOI: 10.3390/pharmaceutics16010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Multiple sclerosis is an important neurological disease affecting millions of young patients globally. It is encouraging that more than ten disease-modifying drugs became available for use in the past two decades. These disease-modifying therapies (DMTs) have different levels of efficacy, routes of administration, adverse effect profiles and concerns for pregnancy. Much knowledge and caution are needed for their appropriate use in MS patients who are heterogeneous in clinical features and severity, lesion load on magnetic resonance imaging and response to DMT. We aim for an updated review of the concept of personalization in the use of DMT for relapsing MS patients. Shared decision making with consideration for the preference and expectation of patients who understand the potential efficacy/benefits and risks of DMT is advocated.
Collapse
Affiliation(s)
- Chi-Yan Lee
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 405B, 4/F, Professorial Block, 102 Pokfulam Road, Hong Kong
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Koon-Ho Chan
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, 405B, 4/F, Professorial Block, 102 Pokfulam Road, Hong Kong
- Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
- Research Center of Heart, Brain, Hormone and Healthy Aging, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
19
|
Broos JY, van der Burgt RTM, Konings J, Rijnsburger M, Werz O, de Vries HE, Giera M, Kooij G. Arachidonic acid-derived lipid mediators in multiple sclerosis pathogenesis: fueling or dampening disease progression? J Neuroinflammation 2024; 21:21. [PMID: 38233951 PMCID: PMC10792915 DOI: 10.1186/s12974-023-02981-w] [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: 09/22/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), characterized by neuroinflammation, demyelination, and neurodegeneration. Considering the increasing prevalence among young adults worldwide and the disabling phenotype of the disease, a deeper understanding of the complexity of the disease pathogenesis is needed to ultimately improve diagnosis and personalize treatment opportunities. Recent findings suggest that bioactive lipid mediators (LM) derived from ω-3/-6 polyunsaturated fatty acids (PUFA), also termed eicosanoids, may contribute to MS pathogenesis. For example, disturbances in LM profiles and especially those derived from the ω-6 PUFA arachidonic acid (AA) have been reported in people with MS (PwMS), where they may contribute to the chronicity of neuroinflammatory processes. Moreover, we have previously shown that certain AA-derived LMs also associated with neurodegenerative processes in PwMS, suggesting that AA-derived LMs are involved in more pathological events than solely neuroinflammation. Yet, to date, a comprehensive overview of the contribution of these LMs to MS-associated pathological processes remains elusive. MAIN BODY This review summarizes and critically evaluates the current body of literature on the eicosanoid biosynthetic pathway and its contribution to key pathological hallmarks of MS during different disease stages. Various parts of the eicosanoid pathway are highlighted, namely, the prostanoid, leukotriene, and hydroxyeicosatetraenoic acids (HETEs) biochemical routes that include specific enzymes of the cyclooxygenases (COXs) and lipoxygenases (LOX) families. In addition, cellular sources of LMs and their potential target cells based on receptor expression profiles will be discussed in the context of MS. Finally, we propose novel therapeutic approaches based on eicosanoid pathway and/or receptor modulation to ultimately target chronic neuroinflammation, demyelination and neurodegeneration in MS. SHORT CONCLUSION The eicosanoid pathway is intrinsically linked to specific aspects of MS pathogenesis. Therefore, we propose that novel intervention strategies, with the aim of accurately modulating the eicosanoid pathway towards the biosynthesis of beneficial LMs, can potentially contribute to more patient- and MS subtype-specific treatment opportunities to combat MS.
Collapse
Affiliation(s)
- Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rianne T M van der Burgt
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
| | - Julia Konings
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Merel Rijnsburger
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Palavra F, Geria L, Jorge A, Marques M, dos Santos CS, Amaral J, Ribeiro JA, Pereira C, Robalo C. Neutrophil/lymphocyte and monocyte/lymphocyte indexes as potential predictors of relapse at 1 year after diagnosis of pediatric multiple sclerosis: a single-center, exploratory and proof-of-concept study. Front Neurosci 2024; 17:1305176. [PMID: 38287987 PMCID: PMC10822923 DOI: 10.3389/fnins.2023.1305176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Early identification of patients with a more unfavorable outcome in Multiple Sclerosis (MS) is crucial to optimize individualized treatment. Neutrophil-lymphocyte index (NLI) and monocyte-lymphocyte index (MLI) have been considered as potential biomarkers for disease prognosis. Our study aims to investigate the usefulness of NLI and MLI as predictors of relapse, disability progression, and lesion accumulation on magnetic resonance imaging (MRI) 1 year after diagnosis and treatment initiation, in pediatric-onset MS. Methods A retrospective single-center study was conducted, including patients with diagnosis of MS established in pediatric age (<18 years old), at least 1-year of follow-up, and a complete blood count (CBC) performed at diagnosis. We collected the nearest-to-diagnosis NLI and MLI, as well as clinical and imaging variables, at diagnosis and 12 months later. Our cohort was further dichotomized into two groups, based on the presence of relapses. Statistical significance was considered for p < 0.05. Results Eighteen patients (n = 18) were included. The relapsing group had higher mean, minimum, and maximum values for both NLI (5.17 ± 5.85, range: 1.57-11.92) and MLI (0.35 ± 0.22, range: 0.19-0.59), compared to the non-relapsing group (2.19 ± 1.63, range: 1.12-7.32 for NLI, and 0.24 ± 0.09, range: 0.14-0.44 for MLI). A higher percentage of patients in the relapsing group had increased NLI (>1.89, 66.7%) and MLI (>0.21, 66.7%) values than those in the non-relapsing group (46.7%). Patients who presented new T2-hyperintense lesions on MRI after 1 year of follow-up also had higher mean, minimum, and maximum values of both biomarkers. Patients who did not achieve No Evidence of Disease Activity-3 (NEDA-3) state exhibited higher values for both ratios. However, in our sample, no statistically significant correlations were found between MLI and NLI values and the clinical and imaging variables considered. Conclusion The ease of obtaining NLI and MLI from routine blood tests renders them useful biomarkers as a screening tool in longitudinal follow-up. Our study was based on a very small sample size, but it allowed us to verify the feasibility of the protocol used. It is intended to involve other centers in the next phase of this work, testing the possible usefulness of the indices under analysis on a larger sample.
Collapse
Affiliation(s)
- Filipe Palavra
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Leonor Geria
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - André Jorge
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Margarida Marques
- Biostatistics and Medical Informatics Laboratory, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Constança Soares dos Santos
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Joana Amaral
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Joana Afonso Ribeiro
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Cristina Pereira
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Conceição Robalo
- Center for Child Development–Neuropediatrics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| |
Collapse
|
21
|
Dou M, Chu Y, Zhou X, Wang M, Li X, Ma R, Fan Z, Zhao X, Wang W, Li S, Lv Y, Zhu L. Matrine Mediated Immune Protection in MS by Regulating Gut Microbiota and Production of SCFAs. Mol Neurobiol 2024; 61:74-90. [PMID: 37581848 DOI: 10.1007/s12035-023-03568-5] [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: 05/16/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
There is clearly an unmet need for more effective and safer treatments for multiple sclerosis (MS). Our previous studies showed a significant therapeutic effect of matrine, a monomer of traditional herbal medicine, on experimental autoimmune encephalomyelitis (EAE) mice. To explore the mechanism of matrine action, we used 16S rRNA sequencing technology to determine the gut microbes in matrine-treated EAE mice and controls. The concentrations of short-chain fatty acids (SCFAs) were then tested by metabonomics. Finally, we established pseudo-sterile mice and transplanted into them fecal microbiota, which had been obtained from the high-dose matrine-treated EAE mice to test the effects of matrine. The results showed that matrine could restore the diversity of gut microbiota and promote the production of SCFAs in EAE mice. Transplantation of fecal microbiota from matrine-treated mice significantly alleviated EAE severity, reduced CNS inflammatory infiltration and demyelination, and decreased the level of IL-17 but increased IL-10 in sera of mice. In conclusion, matrine treatment can regulate gut microbiota and metabolites and halt the progression of MS.
Collapse
Affiliation(s)
- Mengmeng Dou
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Integrated Traditional and Western Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaojuan Chu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xueliang Zhou
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengru Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinyu Li
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Ma
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhirui Fan
- Department of Integrated Traditional and Western Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyu Zhao
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenbin Wang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Silu Li
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Lv
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Zhu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
22
|
Jia Q, Bai D, Zheng X, Zhu L, Ou K, Wang X, Tong H, Zhang Y, Wang J, Zeng J, Yan S, Li S, Li XJ, Yin P. Comparing HD knockin pigs and mice reveals the pathological role of IL-17. Cell Rep 2023; 42:113443. [PMID: 37979175 DOI: 10.1016/j.celrep.2023.113443] [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: 06/14/2023] [Revised: 09/09/2023] [Accepted: 10/30/2023] [Indexed: 11/20/2023] Open
Abstract
Our previous work has established a knockin (KI) pig model of Huntington's disease (HD) that can replicate the typical pathological features of HD, including selective striatal neuronal loss, reactive gliosis, and axonal degeneration. However, HD KI mice exhibit milder neuropathological phenotypes and lack overt neurodegeneration. By performing RNA sequencing to compare the gene expression profiles between HD KI pigs and mice, we find that genes related to interleukin-17 (IL-17) signaling are upregulated in the HD pig brains compared to the mouse brains. Delivery of IL-17 into the brain striatum of HD KI mice causes greater reactive gliosis and synaptic deficiency compared to HD KI mice that received PBS. These findings suggest that the upregulation of genes related to IL-17 signaling in HD pig brains contributes to severe glial pathology in HD and identify this as a potential therapeutic target for treating HD.
Collapse
Affiliation(s)
- Qingqing Jia
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Dazhang Bai
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of Neurological Diseases, North Sichuan Medical College, Nanchong 637000, China
| | - Xiao Zheng
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Longhong Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Kaili Ou
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Xiang Wang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Huichun Tong
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Jing Wang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Jun Zeng
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510260, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
23
|
Hosseinkhani B, Duran G, Hoeks C, Hermans D, Schepers M, Baeten P, Poelmans J, Coenen B, Bekar K, Pintelon I, Timmermans JP, Vanmierlo T, Michiels L, Hellings N, Broux B. Cerebral microvascular endothelial cell-derived extracellular vesicles regulate blood - brain barrier function. Fluids Barriers CNS 2023; 20:95. [PMID: 38114994 PMCID: PMC10729529 DOI: 10.1186/s12987-023-00504-6] [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: 09/07/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023] Open
Abstract
Autoreactive T lymphocytes crossing the blood-brain barrier (BBB) into the central nervous system (CNS) play a crucial role in the initiation of demyelination and neurodegeneration in multiple sclerosis (MS). Recently, extracellular vesicles (EV) secreted by BBB endothelial cells (BBB-EC) have emerged as a unique form of cell-to-cell communication that contributes to cerebrovascular dysfunction. However, the precise impact of different size-based subpopulations of BBB-EC-derived EV (BBB-EV) on the early stages of MS remains unclear. Therefore, our objective was to investigate the content and function of distinct BBB-EV subpopulations in regulating BBB integrity and their role in T cell transendothelial migration, both in vitro and in vivo. Our study reveals that BBB-ECs release two distinct size based EV populations, namely small EV (sEV; 30-150 nm) and large EV (lEV; 150-300 nm), with a significantly higher secretion of sEV during inflammation. Notably, the expression patterns of cytokines and adhesion markers differ significantly between these BBB-EV subsets, indicating specific functional differences in the regulation of T cell migration. Through in vitro experiments, we demonstrate that lEV, which predominantly reflect their cellular source, play a major role in BBB integrity loss and the enhanced migration of pro-inflammatory Th1 and Th17.1 cells. Conversely, sEV appear to protect BBB function by inducing an anti-inflammatory phenotype in BBB-EC. These findings align with our in vivo data, where the administration of sEV to mice with experimental autoimmune encephalomyelitis (EAE) results in lower disease severity compared to the administration of lEV, which exacerbates disease symptoms. In conclusion, our study highlights the distinct and opposing effects of BBB-EV subpopulations on the BBB, both in vitro and in vivo. These findings underscore the need for further investigation into the diagnostic and therapeutic potential of BBB-EV in the context of MS.
Collapse
Affiliation(s)
- Baharak Hosseinkhani
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, KU Leuven, Leuven, Belgium
- Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Gayel Duran
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Cindy Hoeks
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Doryssa Hermans
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Melissa Schepers
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Paulien Baeten
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Joren Poelmans
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Britt Coenen
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Kübra Bekar
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology & Histology/Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Universiteitsplein 1, Antwerp, 2610, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology & Histology/Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Universiteitsplein 1, Antwerp, 2610, Belgium
| | - Tim Vanmierlo
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Luc Michiels
- Bionanotechnology group, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Niels Hellings
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Bieke Broux
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium.
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium.
- Universiteit Hasselt, Martelarenlaan 42, Hasselt, Belgium.
| |
Collapse
|
24
|
Vakrakou AG, Brinia ME, Alexaki A, Koumasopoulos E, Stathopoulos P, Evangelopoulos ME, Stefanis L, Stadelmann-Nessler C, Kilidireas C. Multiple faces of multiple sclerosis in the era of highly efficient treatment modalities: Lymphopenia and switching treatment options challenges daily practice. Int Immunopharmacol 2023; 125:111192. [PMID: 37951198 DOI: 10.1016/j.intimp.2023.111192] [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: 09/03/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
The expanded treatment landscape in relapsing-remitting multiple sclerosis (MS) has resulted in highly effective treatment options and complexity in managing disease- or drug-related events during disease progression. Proper decision-making requires thorough knowledge of the immunobiology of MS itself and an understanding of the main principles behind the mechanisms that lead to secondary autoimmunity affecting organs other than the central nervous system as well as opportunistic infections. The immune system is highly adapted to both environmental and disease-modifying agents. Immune reconstitution following cell depletion or cell entrapment therapies eliminates pathogenic aspects of the disease but can also lead to distorted immune responses with harmful effects. Atypical relapses occur with second-line treatments or after their discontinuation and require appropriate clinical decisions. Lymphopenia is a result of the mechanism of action of many drugs used to treat MS. However, persistent lymphopenia and cell-specific lymphopenia could result in disease exacerbation, secondary autoimmunity, or the emergence of opportunistic infections. Clinicians treating patients with MS should be aware of the multiple faces of MS under novel, efficient treatment modalities and understand the intricate brain-immune cell interactions in the context of an altered immune system. MS relapses and disease progression still occur despite the current treatment modalities and are mediated either by failure to control effector mechanisms inherent to MS pathophysiology or by new drug-related mechanisms. The multiple faces of MS due to the highly adapted immune system of patients impose the need for appropriate switching therapies that safeguard disease remission and further clinical improvement.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany.
| | - Maria-Evgenia Brinia
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Alexaki
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koumasopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panos Stathopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
25
|
Xia Z, Hu B, Yang M, He W. Zinc finger protein 189 promotes the differentiation of lamina propria T helper 17.1 cells in dextran sulfate sodium-induced colitis. Autoimmunity 2023; 56:2189140. [PMID: 36942486 DOI: 10.1080/08916934.2023.2189140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The factors regulating the heterogeneity of interleukin-17A (IL-17A)-expressing CD4+ T cells in inflammatory bowel diseases remain unclear. In the current study, we characterised the expression and function of zinc finger protein 189 (ZFP189) in a murine colitis model. Mice were given dextran sulphate sodium to induce acute colitis. Flow cytometry was applied to recognise and enrich Th17 and Th17.1 cells based on the expression of IL-17A, interferon-γ (IFN-γ), C-X-C motif chemokine receptor 3 (CXCR3), and C-C motif chemokine receptor 4 (CCR4). The expression of ZFP189 in Th17 and Th17.1 cells was determined by Immunoblotting. Lentivirus-mediated ZFP189 knockdown was conducted to evaluate the effect of ZFP189 on the differentiation of Th17 and Th17.1 cells. The adoptive transfer was performed to analyse the pathogenicity of Th17.1 cells in vivo. We found that ZFP189 was mildly up-regulated in IL-17A-expressing CD4+ T cells in colonic lamina propria. Lamina propria Th17.1 cells expressed higher ZFP189 than Th17 cells. In vitro ZFP189 knockdown in CD4+ T cells did not impact Th17 cell differentiation but suppressed Th17.1 cell differentiation, as evidenced by lower T-box expressed in T cells (T-bet) and IFN-γ. When adoptively transferred into mice, ZFP189-deficient Th17.1 cells produced fewer IFN-γ, tumour necrosis factor-alpha (TNF-α), and granulocyte-macrophage colony-stimulating factor (GM-CSF) than ZFP189-expressing Th17.1 cells. Moreover, ZFP189-deficient Th17.1 cells induced less severe colitis than ZFP189-expressing Th17.1 cells, as evidenced by less body weight loss, a lower disease activity index, and a lower colon histological score. In summary, ZFP189 acts as a positive regulator of the differentiation and pathogenicity of lamina propria Th17.1 cells in colitis.
Collapse
Affiliation(s)
- Zhihong Xia
- Department of General Surgery, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Bo Hu
- Department of Gastrointestinal Surgery, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Min Yang
- Department of General Surgery, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| | - Wenjie He
- Department of Gastrointestinal Surgery, Wuhan Fourth Hospital, Wuhan, Hubei Province, China
| |
Collapse
|
26
|
Khantakova JN, Mutovina A, Ayriyants KA, Bondar NP. Th17 Cells, Glucocorticoid Resistance, and Depression. Cells 2023; 12:2749. [PMID: 38067176 PMCID: PMC10706111 DOI: 10.3390/cells12232749] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Depression is a severe mental disorder that disrupts mood and social behavior and is one of the most common neuropsychological symptoms of other somatic diseases. During the study of the disease, a number of theories were put forward (monoamine, inflammatory, vascular theories, etc.), but none of those theories fully explain the pathogenesis of the disease. Steroid resistance is a characteristic feature of depression and can affect not only brain cells but also immune cells. T-helper cells 17 type (Th17) are known for their resistance to the inhibitory effects of glucocorticoids. Unlike the inhibitory effect on other subpopulations of T-helper cells, glucocorticoids can enhance the differentiation of Th17 lymphocytes, their migration to the inflammation, and the production of IL-17A, IL-21, and IL-23 in GC-resistant disease. According to the latest data, in depression, especially the treatment-resistant type, the number of Th17 cells in the blood and the production of IL-17A is increased, which correlates with the severity of the disease. However, there is still a significant gap in knowledge regarding the exact mechanisms by which Th17 cells can influence neuroinflammation in depression. In this review, we discuss the mutual effect of glucocorticoid resistance and Th17 lymphocytes on the pathogenesis of depression.
Collapse
Affiliation(s)
- Julia N. Khantakova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
| | - Anastasia Mutovina
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia;
| | - Kseniya A. Ayriyants
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
| | - Natalia P. Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia;
| |
Collapse
|
27
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Alasmari AF, Shahid M, Al-Mazroua HA, Alomar HA, AsSobeai HM, Alshamrani AA, Attia SM. MAP kinase inhibitor PD98059 regulates Th1, Th9, Th17, and natural T regulatory cells in an experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. Eur J Pharmacol 2023; 959:176086. [PMID: 37832863 DOI: 10.1016/j.ejphar.2023.176086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/09/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
Experimental autoimmune encephalitis (EAE), an animal model of multiple sclerosis (MS), provides significant insights into the mechanisms that initiate and drive autoimmunity. MS is a chronic autoimmune disease of the central nervous system, characterized by inflammatory infiltration associated with demyelination. T lymphocyte cells play a crucial role in MS, whereas natural T regulatory (nTreg) cells prevent autoimmune inflammation by suppressing lymphocyte activity. This study sought to investigate the role of PD98059, a selective MAP kinase inhibitor, in Th1, Th9, Th17, and nTreg cells using the SJL/J mouse model of EAE. Following EAE development, the mice were intraperitoneally administered PD98059 (5 mg/kg for two weeks) daily. We evaluated the effects of PD98059 on Th1 (IFN-γ and T-bet), Th9 (IL-9 and IRF4), Th17 (IL-17A and RORγT), and nTreg (FoxP3 and Helios) cells in the spleen using flow cytometry. Moreover, we explored the effects of PD98059 on the IFN-γ, T-bet, IL-9, IRF4, IL-17A, RORγT, FoxP3, and Helios mRNA and protein levels in brain tissues using qRT-PCR and Western blot analyses. PD98059 treatment significantly decreased the proportion of CD4+IFN-γ+, CD4+T-bet+, CD4+IL-9+, CD4+IRF4+, CD4+IL-17A+, CD4+RORγT+, CD4+IL-17A+, and CD4+RORγT+ cells while increasing that of CD4+FoxP3+ and CD4+Helios+ cells. In addition, PD98059 administration decreased the mRNA and protein levels of IFN-γ, T-bet, IL-9, IRF4, IL-17A, and RORγT but increased those of FoxP3 and Helios in the brain tissue of EAE mice. Our findings suggest that PD98059 corrects immune dysfunction in EAE mice, which is concurrent with the modulation of multiple signaling pathways.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Homood M AsSobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali A Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
28
|
Thomas OG, Olsson T. Mimicking the brain: Epstein-Barr virus and foreign agents as drivers of neuroimmune attack in multiple sclerosis. Front Immunol 2023; 14:1304281. [PMID: 38022632 PMCID: PMC10655090 DOI: 10.3389/fimmu.2023.1304281] [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: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
T cells have an essential role in adaptive immunity against pathogens and cancer, but failure of thymic tolerance mechanisms can instead lead to escape of T cells with the ability to attack host tissues. Multiple sclerosis (MS) occurs when structures such as myelin and neurons in the central nervous system (CNS) are the target of autoreactive immune responses, resulting in lesions in the brain and spinal cord which cause varied and episodic neurological deficits. A role for autoreactive T cell and antibody responses in MS is likely, and mounting evidence implicates Epstein-Barr virus (EBV) in disease mechanisms. In this review we discuss antigen specificity of T cells involved in development and progression of MS. We examine the current evidence that these T cells can target multiple antigens such as those from pathogens including EBV and briefly describe other mechanisms through which viruses could affect disease. Unravelling the complexity of the autoantigen T cell repertoire is essential for understanding key events in the development and progression of MS, with wider implications for development of future therapies.
Collapse
Affiliation(s)
- Olivia G. Thomas
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Neuroimmunology Unit, Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
29
|
Nguyen Ky M, Duran A, Hasantari I, Bru A, Deloire M, Brochet B, Ruet A, Schmitt N. Natalizumab Treatment Induces Proinflammatory CD4 T Cells Preferentially in the Integrin β7+ Compartment. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200166. [PMID: 37739811 PMCID: PMC10519437 DOI: 10.1212/nxi.0000000000200166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/19/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND AND OBJECTIVES Natalizumab, a monoclonal humanized antibody targeting integrin α4, inhibits the transmigration of lymphocytes into the CNS by preventing the interaction of integrin α4β1 with V-CAM expressed on brain vascular endothelial cells. Although natalizumab treatment reduces the clinical relapse rate in patients with relapsing-remitting MS, its discontinuation after reactivation of the JC virus is associated with a rebound of the disease in 20% of patients. The mechanisms of this rebound are not elucidated, but natalizumab increases the frequencies of circulating CD4 T cells expressing proinflammatory cytokines as well as the proportion of circulating Th17/Th1 cells (Th1-like Th17 cells). Gut-derived memory CD4 T cells are a population of growing interest in the pathogenesis of MS, but whether and how their properties are affected by natalizumab is not known. Here, we studied the phenotype and cytokine expression profile of circulating gut-derived memory CD4 T cells in patients with relapsing-remitting MS under natalizumab. METHODS We identified gut-derived memory CD4 T cells by their expression of integrin β7 and compared their properties and those of integrin β7- memory CD4 T cells across healthy donors and patients with relapsing-remitting MS treated or not with natalizumab. We also compared the capacity of integrin β7- and integrin β7+ CD4 T-cell subsets to transmigrate in vitro across a model of blood-brain barrier. RESULTS The proportions of proinflammatory Th17/Th1 cells as well as of IL-17A+IFNγ+ and IL-17A+GM-CSF+ cells were higher in memory CD4 T cells expressing integrin β7 in patients receiving natalizumab compared with healthy donors and patients with relapsing-remitting MS not receiving natalizumab. By contrast, integrin β7 negative memory CD4 T cells only presented a modest increased in their proportion of Th17/Th1 cells under natalizumab. We further observed that integrin β7+ Th17/Th1 cells migrated as efficiently as integrin β7- Th17/Th1 across a monolayer of brain microvascular endothelial cells. DISCUSSION Our study shows that circulating integrin β7+ memory CD4 T cells of patients with relapsing-remitting MS under natalizumab are enriched in proinflammatory cells supporting the hypothesis that integrin β7+ memory CD4 T cells could play a pathogenic role in the disease rebound observed at natalizumab discontinuation.
Collapse
Affiliation(s)
- Mélanie Nguyen Ky
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Adrien Duran
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Iris Hasantari
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Agnès Bru
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Mathilde Deloire
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Bruno Brochet
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Aurélie Ruet
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France
| | - Nathalie Schmitt
- From the Immunoconcept (M.N.K., A.D., I.H., A.B., N.S.), CNRS UMR 5164, University of Bordeaux; Service de Neurologie (M.D., A.R.), CRC SEP, Centre Hospitalier Universitaire (CHU) de Bordeaux; and INSERM U 1215 (B.B., A.R.), Neurocentre Magendie, University of Bordeaux, France.
| |
Collapse
|
30
|
Aldossari AA, Assiri MA, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Albekairi TH, Alomar HA, Al-Mazroua HA, Almanaa TN, Al-Hamamah MA, Alwetaid MY, Ahmad SF. Histamine H4 Receptor Antagonist Ameliorates the Progression of Experimental Autoimmune Encephalomyelitis via Regulation of T-Cell Imbalance. Int J Mol Sci 2023; 24:15273. [PMID: 37894952 PMCID: PMC10607370 DOI: 10.3390/ijms242015273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Multiple sclerosis (MS) is a degenerative condition characterized by immune-mediated attacks on the central nervous system (CNS), resulting in demyelination and recurring T-cell responses. The histamine H4 receptor (H4R) is mainly expressed in cellular populations and plays a vital role in inflammation and immunological responses. The role of H4R in neurons of the CNS has recently been revealed. However, the precise role of H4R in neuronal function remains inadequately understood. The objective of this work was to investigate the impact of JNJ 10191584 (JNJ), a highly effective and specific H4R antagonist, on the development of experimental autoimmune encephalomyelitis (EAE) and to gain insight into the underlying mechanism involved. In this study, we examined the potential impact of JNJ therapy on the course of EAE in SJL/J mice. EAE mice were administered an oral dose of JNJ at a concentration of 6 mg/kg once a day, starting from day 10 and continuing until day 42. Afterward, the mice's clinical scores were assessed. In this study, we conducted additional research to examine the impact of JNJ on several types of immune cells, specifically Th1 (IFN-γ and T-bet), Th9 (IL-9 and IRF4), Th17 (IL-17A and RORγt), and regulatory T (Tregs; Foxp3 and TGF-β1) cells in the spleen. In this study, we further investigated the impact of JNJ on the mRNA expression levels of IFN-γ, T-bet, IL-9, IRF4, IL-17A, RORγt, Foxp3, and TGF-β1 in the brain. Daily treatment of JNJ effectively reduced the development of EAE in mice. The percentages of CD4+IFN-γ+, CD4+T-bet+, CD4+IL-9+, CD4+IRF4+, CD4+IL-17A+, and CD4+RORγt+ cells were shown to decrease, whereas the percentages of CD4+TGF-β1+ and CD4+Foxp3+ cells were observed to increase in EAE mice treated with JNJ. Therefore, the HR4 antagonist positively affected the course of EAE by modulating the signaling of transcription factors. The identified results include possible ramifications in the context of MS treatment.
Collapse
Affiliation(s)
- Abdullah A. Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A. Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hatun A. Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A. Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Taghreed N. Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Al-Hamamah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Y. Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Toghi M, Bitarafan S, Ghafouri-Fard S. Pathogenic Th17 cells in autoimmunity with regard to rheumatoid arthritis. Pathol Res Pract 2023; 250:154818. [PMID: 37729783 DOI: 10.1016/j.prp.2023.154818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Th17 cells contribute the pathobiology of autoimmune diseases, including rheumatoid arthritis (RA). However, it was shown that differentiated Th17 cells display a high degree of plasticity under the influence of inflammatory conditions. In some autoimmune diseases, the majority of Th17 cells, especially at sites of inflammation, have a phenotype that is intermediate between Th17 and Th1. These cells, which are described as Th17.1 or exTh17 cells, are hypothesized to be more pathogenic than classical Th17 cells. In this review, the involvement of Th17.1 lymphocytes in RA, and potential features that might render these cells to be more pathogenic are discussed.
Collapse
Affiliation(s)
- Mehdi Toghi
- Department of Immune and Infectious Diseases, Université Laval, Quebec City, Quebec, Canada
| | - Sara Bitarafan
- Department of Molecular Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Huang Z, Liu Y, An X, Zhang C, Zhang TX, Li H, Feng B, Li YY, Zhang C. Rituximab induces a transient fluctuation of peripheral and follicular helper T cells in neuromyelitis optica spectrum disorder. J Neuroimmunol 2023; 382:578167. [PMID: 37536049 DOI: 10.1016/j.jneuroim.2023.578167] [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: 04/19/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
Autoreactive CD4+ helper T cells are implicated in the pathogenesis of neuromyelitis optica spectrum disorder (NMOSD). Both PD-1+CXCR5+CD4+ T follicular helper (Tfh) cells and PD-1+CXCR5-CD4+ T peripheral helper (Tph) cells can contribute to B-cell immune responses and the production of antibodies. Here we show the effect of B-cell depletion with rituximab on the homeostasis of Tfh cells, Tph cells and their subsets in patients with NMOSD. After rituximab treatment, total Tph cells, total Tfh cells, Tph17 cells, Tph17.1 cells, Tph1 cells, and Tfh1 cells tended to decrease at month 1, but gradually increased at month 6 and restored at month 12. Besides, Tph17.1 cells and Tfh17.1 cells were correlated with the proportion of CD19- antibody-secreting cells. Our data suggest that rituximab induced a fluctuation of proinflammatory Tph and Tfh subsets within one year after initiation of the treatment.
Collapse
Affiliation(s)
- Zhenning Huang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ye Liu
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueting An
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tian-Xiang Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huining Li
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Feng
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-Yan Li
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China; Center of Neuroimmunology and Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
33
|
Matsuzaka Y, Yashiro R. Unraveling the Immunopathogenesis of Multiple Sclerosis: The Dynamic Dance of Plasmablasts and Pathogenic T Cells. BIOLOGICS 2023; 3:232-252. [DOI: 10.3390/biologics3030013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, characterized by multiple lesions occurring temporally and spatially. Additionally, MS is a disease that predominates in the white population. In recent years, there has been a rapid increase in the number of patients, and it often occurs in young people, with an average age of onset of around 30 years old, but it can also occur in children and the elderly. It is more common in women than men, with a male-to-female ratio of approximately 1:3. As the immunopathogenesis of MS, a group of B cells called plasmablasts controls encephalomyelitis via IL-10 production. These IL-10-producing B cells, called regulatory B cells, suppress inflammatory responses in experimental mouse models of autoimmune diseases including MS. Since it has been clarified that these regulatory B cells are plasmablasts, it is expected that the artificial control of plasmablast differentiation will lead to the development of new treatments for MS. Among CD8-positive T cells in the peripheral blood, the proportion of PD-1-positive cells is decreased in MS patients compared with healthy controls. The dysfunction of inhibitory receptors expressed on T cells is known to be the core of MS immunopathology and may be the cause of chronic persistent inflammation. The PD-1+ CD8+ T cells may also serve as indicators that reflect the condition of each patient in other immunological neurological diseases such as MS. Th17 cells also regulate the development of various autoimmune diseases, including MS. Thus, the restoration of weakened immune regulatory functions may be a true disease-modifying treatment. So far, steroids and immunosuppressants have been the mainstream for autoimmune diseases, but the problem is that this kills not only pathogenic T cells, but also lymphocytes, which are necessary for the body. From this understanding of the immune regulation of MS, we can expect the development of therapeutic strategies that target only pathogenic immune cells.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ryu Yashiro
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
34
|
von Essen MR, Chow HH, Holm Hansen R, Buhelt S, Sellebjerg F. Immune reconstitution following alemtuzumab therapy is characterized by exhausted T cells, increased regulatory control of proinflammatory T cells and reduced B cell control. Front Immunol 2023; 14:1249201. [PMID: 37744364 PMCID: PMC10512074 DOI: 10.3389/fimmu.2023.1249201] [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: 06/28/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Alemtuzumab is a monoclonal antibody targeting CD52 on the surface of immune cells, approved for the treatment of active relapsing-remitting multiple sclerosis (RRMS). The purpose of this study was to analyze the repopulation of peripheral lymphocytes following alemtuzumab-induced lymphocyte depletion and investigate associations with disease activity and development of secondary autoimmunity. For this, blood samples were collected two years after initiation of alemtuzumab treatment and lymphocytes were subjected to a comprehensive flow cytometry analysis. Included in the study were 40 patients treated with alemtuzumab and 40 treatment-naïve patients with RRMS. Disease activity and development of secondary autoimmune disease was evaluated after three years of treatment. Our study confirms that alemtuzumab treatment profoundly alters the circulating lymphocyte phenotype and describes a reconstituted immune system characterized by T cell activation/exhaustion, an increased regulatory control of IL-17 producing effector T cells and CD20+ T cells, and a reduced control of B cells. There were no obvious associations between immune cell subsets and disease activity or development of secondary autoimmune disease during treatment with alemtuzumab. Our results indicate that the reconstituted immune response is skewed towards a more effective regulatory control of MS-associated proinflammatory T cell responses. Also, the enlarged pool of naïve B cells together with the apparent decrease in control of B cell activity may explain why alemtuzumab-treated patients retain the ability to mount a humoral immune response towards new antigens.
Collapse
Affiliation(s)
- Marina Rode von Essen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | | | | | | | | |
Collapse
|
35
|
Holm Hansen R, von Essen MR, Mahler MR, Cobanovic S, Binko TS, Sellebjerg F. Cladribine Effects on T and B Cells and T Cell Reactivity in Multiple Sclerosis. Ann Neurol 2023; 94:518-530. [PMID: 37191113 DOI: 10.1002/ana.26684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Cladribine tablet therapy is an efficacious treatment for multiple sclerosis (MS), however, its mechanism of action on T and B cell subsets remains unclear. The purpose of this study was to investigate the treatment effects of cladribine on the peripheral pool of T and B cells subsets and reactivity toward central nervous system (CNS) antigens. METHODS In this cross-sectional exploratory study, frequencies and absolute counts of peripheral T and B cell subsets and B cell cytokine production from untreated patients with relapsing-remitting MS (RRMS) and patients treated with cladribine for 1 year were measured using flow cytometry. Autoreactivity was assessed using a FluoroSpot assay. RESULTS We found that 1 year after initiation of cladribine treatment, a lower number of CD4+ T cells was persisting whereas CD19+ B cell counts were normalized compared to untreated patients with RRMS. Follicular helper T cells and their effecter subsets producing cytokines exerting distinct B cell helper activity were lower and, additionally, the peripheral B cell pool was skewed toward a naïve and anti-inflammatory phenotype. Finally, reactivity to the recently identified CNS-enriched autoantigen RAS guanyl-releasing protein 2 (RASGRP2), but not to myelin basic protein and myelin oligodendrocyte glycoprotein, was lower in cladribine-treated patients. INTERPRETATION Together, these investigations on T and B cell subsets suggest that cladribine treatment impairs the B-T cell crosstalk and reduces their ability to mediate pathogenic effector functions. This may result in specific reduction of autoreactivity to RASGRP2 which is expressed in B cells and brain tissue. ANN NEUROL 2023;94:518-530.
Collapse
Affiliation(s)
- Rikke Holm Hansen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Marina Rode von Essen
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Mie Reith Mahler
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Stefan Cobanovic
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Tomas Sorm Binko
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Department of Neurology, Danish Multiple Sclerosis Center, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Bozovic I, Perovic V, Basta I, Peric S, Stevic Z, Popadic D, Vukovic I, Stojanov A, Milosevic E. Cytokine Gene Polymorphisms in Patients with Chronic Inflammatory Demyelinating Polyneuropathy. Cells 2023; 12:2033. [PMID: 37626843 PMCID: PMC10453148 DOI: 10.3390/cells12162033] [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: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Innate and adaptive immune responses exert their role in CIDP pathogenesis through cytokine production. Single-nucleotide polymorphisms (SNPs) may alter cytokine gene expression, with a potential influence on the pathogenesis of autoimmune diseases. However, cytokine gene SNPs have not been assessed in CIDP patients yet. We assessed functional SNPs in the genes encoding IL-10 (rs1800896, rs1800871, rs1800872 and rs3024505), IL-6 (rs1800795), TNF (rs1800629 and rs361525), IL-12B (rs3212227), IFN-γ (rs2430561), GM-CSF (rs25882) and IL-17F (rs11465553) in a cohort of 88 CIDP patients and 486 healthy controls (HCs) via qPCR. We found an association of SNP in the IL10 promotor and CIDP occurrence. Major homozygotes (AA) were more frequent in the HCs compared to CIDP patients (p = 0.049), but the GA genotype prevailed among the patients (p = 0.032). A lower frequency of the C allele was observed for rs1800871 and rs1800872 in CIDP patients compared to the HCs (p = 0.048). A higher proportion of A carriers at position -1082 (rs1800896) (presumed to be a low IL-10 producer) was noted in patients with milder disability (low INCAT). All mild-INCAT patients were C carriers for rs1800871 and rs1800872 in IL10 (p = 0.038). Furthermore, the IL6 rs1800795 GG genotype was more frequent in patients (p = 0.049) and the CG heterozygote in the HCs (p = 0.013). Among the CIDP patients, being a G carrier for this SNP was associated with a higher frequency of type 2 diabetes (T2D) compared to being a non-carrier (p = 0.032). Our data indicate a possible association of the IL10 and IL6 SNPs with CIDP, but also with disease severity and T2D occurrence. Given the paucity of CIDP patients, multicentric studies are necessary to draw definite conclusions on these associations.
Collapse
Affiliation(s)
- Ivo Bozovic
- Neurology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | - Ivana Basta
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Stojan Peric
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Zorica Stevic
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Dusan Popadic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | - Irena Vukovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | | | - Emina Milosevic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| |
Collapse
|
37
|
Qi C, Feng Y, Jiang Y, Chen W, Vakal S, Chen JF, Zheng W. A 2AR antagonist treatment for multiple sclerosis: Current progress and future prospects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:185-223. [PMID: 37741692 DOI: 10.1016/bs.irn.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Emerging evidence suggests that both selective and non-selective Adenosine A2A receptor (A2AR) antagonists could effectively protect mice from experimental autoimmune encephalomyelitis (EAE), which is the most commonly used animal model for multiple sclerosis (MS) research. Meanwhile, the recent FDA approval of Nourianz® (istradefylline) in 2019 as an add-on treatment to levodopa in Parkinson's disease (PD) with "OFF" episodes, along with its proven clinical safety, has prompted us to explore the potential of A2AR antagonists in treating multiple sclerosis (MS) through clinical trials. However, despite promising findings in experimental autoimmune encephalomyelitis (EAE), the complex and contradictory role of A2AR signaling in EAE pathology has raised concerns about the feasibility of using A2AR antagonists as a therapeutic approach for MS. This review addresses the potential effect of A2AR antagonists on EAE/MS in both the peripheral immune system (PIS) and the central nervous system (CNS). In brief, A2AR antagonists had a moderate effect on the proliferation and inflammatory response, while exhibiting a potent anti-inflammatory effect in the CNS through their impact on microglia, astrocytes, and the endothelial cells/epithelium of the blood-brain barrier. Consequently, A2AR signaling remains an essential immunomodulator in EAE/MS, suggesting that A2AR antagonists hold promise as a drug class for treating MS.
Collapse
Affiliation(s)
- Chenxing Qi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yijia Feng
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Yiwei Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou, P.R. China
| | - Wangchao Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Serhii Vakal
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Jiang-Fan Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Wu Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China.
| |
Collapse
|
38
|
Soldati S, Bär A, Vladymyrov M, Glavin D, McGrath JL, Gosselet F, Nishihara H, Goelz S, Engelhardt B. High levels of endothelial ICAM-1 prohibit natalizumab mediated abrogation of CD4 + T cell arrest on the inflamed BBB under flow in vitro. J Neuroinflammation 2023; 20:123. [PMID: 37221552 DOI: 10.1186/s12974-023-02797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/02/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION The humanized anti-α4 integrin blocking antibody natalizumab (NTZ) is an effective treatment for relapsing-remitting multiple sclerosis (RRMS) that is associated with the risk of progressive multifocal leukoencephalopathy (PML). While extended interval dosing (EID) of NTZ reduces the risk for PML, the minimal dose of NTZ required to maintain its therapeutic efficacy remains unknown. OBJECTIVE Here we aimed to identify the minimal NTZ concentration required to inhibit the arrest of human effector/memory CD4+ T cell subsets or of PBMCs to the blood-brain barrier (BBB) under physiological flow in vitro. RESULTS Making use of three different human in vitro BBB models and in vitro live-cell imaging we observed that NTZ mediated inhibition of α4-integrins failed to abrogate T cell arrest to the inflamed BBB under physiological flow. Complete inhibition of shear resistant T cell arrest required additional inhibition of β2-integrins, which correlated with a strong upregulation of endothelial intercellular adhesion molecule (ICAM)-1 on the respective BBB models investigated. Indeed, NTZ mediated inhibition of shear resistant T cell arrest to combinations of immobilized recombinant vascular cell adhesion molecule (VCAM)-1 and ICAM-1 was abrogated in the presence of tenfold higher molar concentrations of ICAM-1 over VCAM-1. Also, monovalent NTZ was less potent than bivalent NTZ in inhibiting T cell arrest to VCAM-1 under physiological flow. In accordance with our previous observations ICAM-1 but not VCAM-1 mediated T cell crawling against the direction of flow. CONCLUSION Taken together, our in vitro observations show that high levels of endothelial ICAM-1 abrogate NTZ mediated inhibition of T cell interaction with the BBB. EID of NTZ in MS patients may thus require consideration of the inflammatory status of the BBB as high levels of ICAM-1 may provide an alternative molecular cue allowing for pathogenic T cell entry into the CNS in the presence of NTZ.
Collapse
Affiliation(s)
- Sasha Soldati
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Alexander Bär
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Mykhailo Vladymyrov
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Dale Glavin
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, University of Artois, Lens, France
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
- Department of Neurotherapeutics, Yamaguchi University, Yamaguchi, Japan
| | | | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland.
| |
Collapse
|
39
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Al-Mazroua HA, Alomar HA, Al-Hamamah MA, Attia SM. S3I-201, a selective stat3 inhibitor, ameliorates clinical symptoms in a mouse model of experimental autoimmune encephalomyelitis through the regulation of multiple intracellular signalling in Th1, Th17, and treg cells. Mult Scler Relat Disord 2023; 73:104658. [PMID: 36989705 DOI: 10.1016/j.msard.2023.104658] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
CD4+ T cells, specifically Th cells (Th1 and Th17) and regulatory T cells (Tregs), play a pivotal role in the pathogenesis of multiple sclerosis (MS), a demyelinating autoimmune disease of the CNS. STAT3 inhibitors are potential therapeutic targets for several immune disorders. In this study, we investigated the role of a well-known STAT3 inhibitor, S3I-201, in experimental autoimmune encephalomyelitis (EAE), a model of MS. Following induction of EAE, mice were intraperitoneally administered S3I-201 (10 mg/kg) each day, beginning on day 14 and continuing till day 35 and were evaluated for clinical signs. Flow cytometry was used to investigate further the effect of S3I-201 on Th1 (IFN-γ, STAT1, pSTAT1, and T-bet), Th17 (IL-17A, STAT3, pSTAT3, and RORγt), and regulatory T cells (Treg, IL-10, TGF-β1, and FoxP3) expressed in splenic CD4+ T cells. Moreover, we analyzed the effects of S3I-201 on mRNA and protein expression of IFN-γ, T-bet, IL-17A, STAT1, STAT3, pSTAT1, pSTAT3, RORγ, IL-10, TGF-β1, and FoxP3 in the brains of EAE mice. The severity of clinical scores decreased in S3I-201-treated EAE mice compared to vehicle-treated EAE mice. S3I-201 treatment significantly decreased CD4+IFN-γ+, CD4+STAT1+, CD4+pSTAT1+, CD4+T-bet+, CD4+IL-17A+, CD4+STAT3+, CD4+pSTAT3+, and CD4+RORγt+ and increased CD4+IL-10+, CD4+TGF-β1+, and CD4+FoxP3+ in the spleens of EAE mice. Additionally, S3I-201 administration in EAE mice significantly decreased the mRNA and protein expression of Th1 and Th17 and increased those of Treg. These results suggest that S3I-201 may have novel therapeutic potential against MS.
Collapse
|
40
|
Mannion JM, McLoughlin RM, Lalor SJ. The Airway Microbiome-IL-17 Axis: a Critical Regulator of Chronic Inflammatory Disease. Clin Rev Allergy Immunol 2023; 64:161-178. [PMID: 35275333 PMCID: PMC10017631 DOI: 10.1007/s12016-022-08928-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 02/07/2023]
Abstract
The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.
Collapse
Affiliation(s)
- Jenny M Mannion
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Stephen J Lalor
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
41
|
Lin LY, Juillard P, Hawke S, Marsh-Wakefield F, Grau GE. Oral Cladribine Impairs Intermediate, but Not Conventional, Monocyte Transmigration in Multiple Sclerosis Patients across a Model Blood-Brain Barrier. Int J Mol Sci 2023; 24:ijms24076487. [PMID: 37047460 PMCID: PMC10094666 DOI: 10.3390/ijms24076487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
Multiple sclerosis (MS) is a disease in which the immune system damages components of the central nervous system (CNS), leading to the destruction of myelin and the formation of demyelinating plaques. This often occurs in episodic “attacks” precipitated by the transmigration of leukocytes across the blood-brain barrier (BBB), and repeated episodes of demyelination lead to substantial losses of axons within and removed from plaques, ultimately leading to progressive neurological dysfunction. Within leukocyte populations, macrophages and T and B lymphocytes are the predominant effectors. Among current immunotherapies, oral cladribine’s impact on lymphocytes is well characterised, but little is known about its impact on other leukocytes such as monocytes and dendritic cells (DCs). The aim of this study was to determine the transmigratory ability of monocyte and DC subsets in healthy subjects and untreated and cladribine-treated relapse-remitting MS (RRMS) patients using a well-characterised model of the BBB. Peripheral blood mononuclear cells from subjects were added to an in vitro transmigration assay to assess cell migration. Our findings show that while prior treatment with oral cladribine inhibits the migration of intermediate monocytes, it has no impact on the transmigration of DC subsets. Overall, our data indicate a previously unrecognised role of cladribine on intermediate monocytes, known to accumulate in the brain active MS lesions.
Collapse
Affiliation(s)
- Linda Y. Lin
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pierre Juillard
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Simon Hawke
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Central West Neurology and Neurosurgery, Orange, NSW 2800, Australia
| | - Felix Marsh-Wakefield
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Liver Injury and Cancer Program, Centenary Institute, Sydney, NSW 2006, Australia
- Human Cancer and Viral Immunology Laboratory, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: (F.M.-W.); (G.E.G.)
| | - Georges E. Grau
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: (F.M.-W.); (G.E.G.)
| |
Collapse
|
42
|
Shafiei M, Mozhgani SH. Th17/IL-17 Axis in HTLV-1-Associated Myelopathy Tropical Spastic Paraparesis and Multiple Sclerosis: Novel Insights into the Immunity During HAMTSP. Mol Neurobiol 2023; 60:3839-3854. [PMID: 36947318 DOI: 10.1007/s12035-023-03303-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
Human T lymphotropic virus-associated myelopathy/tropical spastic paraparesis (HTLV/TSP), also known as HTLV-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and multiple sclerosis (MS) are chronic debilitating diseases of the central nervous system; although the etiology of which is different, similarities have been observed between these two demyelinating diseases, especially in clinical manifestation and immunopathogenesis. Exorbitant response of the immune system to the virus and neurons in CNS is the causative agent of HAM/TSP and MS, respectively. Helper T lymphocyte-17 cells (Th17s), a component of the immune system, which have a proven role in immunity and autoimmunity, mediate protection against bacterial/fungal infections. The role of these cells has been reviewed in several CNS diseases. A pivotal role for Th17s is presented in demyelination, even more axial than Th1s, during MS. The effect of Th17s is not well determined in HTLV-1-associated infections; however, the evidence that we have supplied in this review illustrates the attendance, also the role of Th17 cells during HAM/TSP. Furthermore, for better conception concerning the trace of these cells in HAM/TSP, a comparative characterization with MS, the resembling disease, has been applied here.
Collapse
Affiliation(s)
- Mohammadreza Shafiei
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
43
|
Th17.1 cell driven sarcoidosis-like inflammation after anti-BCMA CAR T cells in multiple myeloma. Leukemia 2023; 37:650-658. [PMID: 36720972 PMCID: PMC9888347 DOI: 10.1038/s41375-023-01824-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023]
Abstract
Pseudo-progression and flare-up phenomena constitute a novel diagnostic challenge in the follow-up of patients treated with immune-oncology drugs. We present a case study on pulmonary flare-up after Idecabtagen Vicleucel (Ide-cel), a BCMA targeting CAR T-cell therapy, and used single-cell RNA-seq (scRNA-seq) to identify a Th17.1 driven autoimmune mechanism as the biological underpinning of this phenomenon. By integrating datasets of various lung pathological conditions, we revealed transcriptomic similarities between post CAR T pulmonary lesions and sarcoidosis. Furthermore, we explored a noninvasive PET based diagnostic approach and showed that tracers binding to CXCR4 complement FDG PET imaging in this setting, allowing discrimination between immune-mediated changes and true relapse after CAR T-cell treatment. In conclusion, our study highlights a Th17.1 driven autoimmune phenomenon after CAR T, which may be misinterpreted as disease relapse, and that imaging with multiple PET tracers and scRNA-seq could help in this diagnostic dilemma.
Collapse
|
44
|
Garcia A, Dugast E, Shah S, Morille J, Lebrun-Frenay C, Thouvenot E, De Sèze J, Le Page E, Vukusic S, Maurousset A, Berger E, Casez O, Labauge P, Ruet A, Raposo C, Bakdache F, Buffels R, Le Frère F, Nicot A, Wiertlewski S, Gourraud PA, Berthelot L, Laplaud D. Immune Profiling Reveals the T-Cell Effect of Ocrelizumab in Early Relapsing-Remitting Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200091. [PMID: 36810163 PMCID: PMC9944617 DOI: 10.1212/nxi.0000000000200091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/12/2022] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND OBJECTIVES Ocrelizumab (OCR), a humanized anti-CD20 monoclonal antibody, is highly efficient in patients with relapsing-remitting multiple sclerosis (RR-MS). We assessed early cellular immune profiles and their association with disease activity at treatment start and under therapy, which may provide new clues on the mechanisms of action of OCR and on the disease pathophysiology. METHODS A first group of 42 patients with an early RR-MS, never exposed to disease-modifying therapy, was included in 11 centers participating to an ancillary study of the ENSEMBLE trial (NCT03085810) to evaluate the effectiveness and safety of OCR. The phenotypic immune profile was comprehensively assessed by multiparametric spectral flow cytometry at baseline and after 24 and 48 weeks of OCR treatment on cryopreserved peripheral blood mononuclear cells and analyzed in relation to disease clinical activity. A second group of 13 untreated patients with RR-MS was included for comparative analysis of peripheral blood and CSF. The transcriptomic profile was assessed by single-cell qPCRs of 96 genes of immunologic interest. RESULTS Using an unbiased analysis, we found that OCR as an effect on 4 clusters of CD4+ T cells: one corresponding to naive CD4+ T cells was increased, the other clusters corresponded to effector memory (EM) CD4+CCR6- T cells expressing homing and migration markers, 2 of them also expressing CCR5 and were decreased by the treatment. Of interest, one CD8+ T-cell cluster was decreased by OCR corresponding to EM CCR5-expressing T cells with high expression of the brain homing markers CD49d and CD11a and correlated with the time elapsed since the last relapse. These EM CD8+CCR5+ T cells were enriched in the CSF of patients with RR-MS and corresponded to activated and cytotoxic cells. DISCUSSION Our study provides novel insights into the mode of action of anti-CD20, pointing toward the role of EM T cells, particularly a subset of CD8 T cells expressing CCR5.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David Laplaud
- From the CHU Nantes (A.G., E.D., S.S., J.M., A.N., S.W., P.-A.G., L.B., D.L.), Nantes Université, INSERM UMR1064, Center for Research in Transplantation and Translational Immunology (CR2TI); CRCSEP (C.L.-F.), CHU de Nice Pasteur 2, Université Nice Côte d'Azur UR2CA URRIS; Service de Neurologie (E.T.), CHU de Nîmes, Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM; Service de Neurologie et Centre d'Investigation Clinique (J.D.S.), CHU de Strasbourg; Service de Neurologie (E.L.P.), CHU Pontchaillou, Rennes; Université de Lyon (S.V.), Université Claude Bernard Lyon 1; Service de Neurologie (S.V.), sclérose en plaques, pathologies de la Myéline et Neuro-inflammation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron; Observatoire Français de la Sclérose en Plaques (S.V.), Centre de Recherche en Neurosciences de Lyon; EUGENE DEVIC EDMUS Foundation Against Multiple sclerosis (S.V.), state-approved Foundation, Bron; Service de Neurologie (A.M.), CHU Bretonneau, Tours; Service de Neurologie (E.B.), CHU de Besançon; Service de Neurologie (O.C.), CHU de Grenoble; Service de Neurologie (P.L.), CHU de Montpellier, Montpellier; Service de Neurologie (A.R.), CHU de Bordeaux; Université de Bordeaux (A.R.), INSERM, Neurocentre Magendie; F. Hoffmann-La Roche Ltd (C.R., F.B., R.B.) CIC INSERM 1413 (F.L.F., S.W., D.L.), Nantes; CHU Nantes (S.W., D.L.), Nantes Université, Service de Neurologie; and CHU Nantes (P.-A.G.), Nantes Université, Clinique des données, France.
| |
Collapse
|
45
|
Pham HPT, Saroukhani S, Lindsey JW. The concentrations of antibodies to Epstein-Barr virus decrease during ocrelizumab treatment. Mult Scler Relat Disord 2023; 70:104497. [PMID: 36603289 DOI: 10.1016/j.msard.2023.104497] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/04/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023]
Abstract
BACKGROUND Epstein-Barr Virus (EBV) is strongly associated with multiple sclerosis (MS). After initial infection, EBV maintains a life-long latent infection in B lymphocytes. Depletion of B lymphocytes from the blood with the anti-CD20 antibody ocrelizumab (OCR) markedly reduces disease activity in MS. Our objective was to measure the effect of OCR treatment on the antibody response to EBV and human antigens that are cross-reactive with EBV. METHODS Blood was collected from MS patients before and during OCR treatment. Antibodies to three EBV antigens (EBNA-1, BFRF3, and gp350) and three human proteins that are cross-reactive with EBV (septin-9, DLST, and HNRNPL) were quantified with Western blots. Antibodies to EBNA-1 and BFRF3 were also quantified with ELISA. RESULTS Antibodies to the EBV proteins BFRF3 and EBNA-1 measured on Western blot were significantly decreased after 12 months on OCR. Subsequent testing with ELISA confirmed the decrease for both BFRF3 and EBNA-1. With Western blots, there was a trend to decreased antibody response to septin-9 and DLST, but not HNRNPL. Total IgG concentration did not change. CONCLUSION The antibody response to some EBV antigens decreases in OCR treated patients. The benefit of OCR for MS may be through removal of EBV antigenic stimulus.
Collapse
Affiliation(s)
- H Phuong T Pham
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sepideh Saroukhani
- Division of Clinical and Translational Sciences, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA; Biostatistics/Epidemiology/Research Design (BERD) component, Center for Clinical and Translational Sciences (CCTS), The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - J William Lindsey
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
46
|
Zamorina S, Timganova V, Bochkova M, Shardina K, Uzhviyuk S, Khramtsov P, Usanina D, Rayev M. The Effect of PEGylated Graphene Oxide Nanoparticles on the Th17-Polarization of Activated T Helpers. MATERIALS (BASEL, SWITZERLAND) 2023; 16:877. [PMID: 36676614 PMCID: PMC9865146 DOI: 10.3390/ma16020877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
We investigated the direct effect of PEGylated graphene oxide (P-GO) nanoparticles on the differentiation, viability, and cytokine profile of activated T helper type 17 (Th17) in vitro. The subject of the study were cultures of "naive" T-helpers (CD4+) isolated by immunomagnetic separation and polarized into the Th17 phenotype with a TCR activator and cytokines. It was found that P-GO at low concentrations (5 µg/mL) had no effect on the parameters studied. The presence of high concentrations of P-GO in T-helper cultures (25 μg/mL) did not affect the number and viability of these cells. However, the percentage of proliferating T-helpers in these cultures was reduced. GO nanoparticles modified with linear polyethylene glycol (PEG) significantly increased the percentage of Th17/22 cells in cultures of Th17-polarized T helpers and the production of IFN-γ, whereas those modified with branched PEG suppressed the synthesis of IL-17. Thus, a low concentration of PEGylated GO nanoparticles (5 μg/mL), in contrast to a concentration of 25 μg/mL, has no effect on the Th17-polarization of T helpers, allowing their further use for in-depth studies of the functions of T lymphocytes and other immune cells. Overall, we have studied for the first time the direct effect of P-GO nanoparticles on the conversion of T helper cells to the Th17 phenotype.
Collapse
Affiliation(s)
- Svetlana Zamorina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Valeria Timganova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Maria Bochkova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Kseniya Shardina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Sofya Uzhviyuk
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Pavel Khramtsov
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Darya Usanina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Mikhail Rayev
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| |
Collapse
|
47
|
de Sèze J, Maillart E, Gueguen A, Laplaud DA, Michel L, Thouvenot E, Zephir H, Zimmer L, Biotti D, Liblau R. Anti-CD20 therapies in multiple sclerosis: From pathology to the clinic. Front Immunol 2023; 14:1004795. [PMID: 37033984 PMCID: PMC10076836 DOI: 10.3389/fimmu.2023.1004795] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system plays a significant role in multiple sclerosis. While MS was historically thought to be T cell-mediated, multiple pieces of evidence now support the view that B cells are essential players in multiple sclerosis pathogenic processes. High-efficacy disease-modifying therapies that target the immune system have emerged over the past two decades. Anti-CD20 monoclonal antibodies selectively deplete CD20+ B and CD20+ T cells and efficiently suppress inflammatory disease activity. These monotherapies prevent relapses, reduce new or active magnetic resonance imaging brain lesions, and lessen disability progression in patients with relapsing multiple sclerosis. Rituximab, ocrelizumab, and ofatumumab are currently used in clinical practice, while phase III clinical trials for ublituximab have been recently completed. In this review, we compare the four anti-CD20 antibodies in terms of their mechanisms of action, routes of administration, immunological targets, and pharmacokinetic properties. A deeper understanding of the individual properties of these molecules in relation to their efficacy and safety profiles is critical for their use in clinical practice.
Collapse
Affiliation(s)
- Jérôme de Sèze
- Department of Neurology, Hôpital de Hautepierre, Clinical Investigation Center, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France
- Fédération de Médecine Translationelle, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France
- *Correspondence: Jérôme de Sèze,
| | - Elisabeth Maillart
- Department of Neurology, Pitié Salpêtrière Hospital, Paris, France
- Centre de Ressources et de Compétences Sclérose en Plaques, Paris, France
| | - Antoine Gueguen
- Department of Neurology, Rothschild Ophthalmologic Foundation, Paris, France
| | - David A. Laplaud
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Nantes, Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Investigation Clinique (CIC), Center for Research in Transplantation and Translational Immunology, UMR, UMR1064, Nantes, France
| | - Laure Michel
- Clinical Neuroscience Centre, CIC_P1414 Institut National de la Santé et de la Recherche Médicale (INSERM), Rennes University Hospital, Rennes University, Rennes, France
- Microenvironment, Cell Differentiation, Immunology and Cancer Unit, Institut National de la Santé et de la Recherche Médicale (INSERM), Rennes I University, French Blood Agency, Rennes, France
- Neurology Department, Rennes University Hospital, Rennes, France
| | - Eric Thouvenot
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Nîmes, University of Montpellier, Nîmes, France
- Institut de Génomique Fonctionnelle, UMR, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Montpellier, Montpellier, France
| | - Hélène Zephir
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM) U1172, Centre Hospitalier Universitaire (CHU), Lille, France
| | - Luc Zimmer
- Université Claude Bernard Lyon 1, Hospices Civils de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS, Lyon Neuroscience Research Center, Lyon, France
| | - Damien Biotti
- Centre Ressources et Compétences Sclérose En Plaques (CRC-SEP) and Department of Neurology, Centre Hospitalier Universitaire (CHU) Toulouse Purpan – Hôpital Pierre-Paul Riquet, Toulouse, France
| | - Roland Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, Institut National de la Santé et de la Recherche Médicale (INSERM), UPS, Toulouse, France
- Department of Immunology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
48
|
Tredicine M, Ria F, Poerio N, Lucchini M, Bianco A, De Santis F, Valentini M, De Arcangelis V, Rende M, Stabile AM, Pistilli A, Camponeschi C, Nociti V, Mirabella M, Fraziano M, Di Sante G. Liposome-based nanoparticles impact on regulatory and effector phenotypes of macrophages and T cells in multiple Sclerosis patients. Biomaterials 2023; 292:121930. [PMID: 36493716 DOI: 10.1016/j.biomaterials.2022.121930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Current available treatments of Multiple Sclerosis (MS) reduce neuroinflammation acting on different targets on the immune system, but potentially lead to severe side effects and have a limited efficacy in slowing the progression of the disease. Here, we evaluated in vitro the immunomodulatory potential of a new class of nanoparticles - liposomes, constituted by a double-layer of phosphatidylserine (PSCho/PS), and double-faced, with an outer layer of phosphatidylserine and an inner layer of phosphatidic acid (PSCho/PA), either alone or in the presence of the myelin basic protein (MBP) peptide (residues 85-99) (PSCho/PS-MBP and PSCho/PA-MBP). Results showed that PSCho/PS are equally and efficiently internalized by pro- and anti-inflammatory macrophages (M1 and M2 respectively), while PSCho/PA were internalized better by M2 than M1. PSCho/PS liposomes were able to inhibit the secretion of innate pro-inflammatory cytokine IL-1β. PSCho/PS liposomes expanded Tregs, reducing Th1 and Th17 cells, while PSCho/PA liposomes were unable to dampen pro-inflammatory T cells and to promote immune-regulatory phenotype (Treg). The ability of PSCho/PS liposomes to up-regulate Treg cells was more pronounced in MS patients with high basal expression of M2 markers. PSCho/PS liposomes were more effective in decreasing Th1 (but not Th17) cells in MS patients with a disease duration >3 months. On the other hand, down-modulation of Th17 cells was evident in MS patients with active, Gadolinium enhancing lesions at MRI and in MS patients with a high basal expression of M1-associated markers in the monocytes. The same findings were observed for the modulation of MBP-driven Th1/Th17/Treg responses. These observations suggest that early MS associate to a hard-wired pro-Th1 phenotype of M1 that is lost later during disease course. On the other hand, acute inflammatory events reflect a temporary decrease of M2 phenotype that however is amenable to restauration upon treatment with PSCho/PS liposomes. Thus, together these data indicate that monocytes/macrophages may play an important regulatory function during MS course and suggest a role for PSCho/PS and PSCho/PS-MBP as new therapeutic tools to dampen the pro-inflammatory immune responses and to promote its regulatory branch.
Collapse
Affiliation(s)
- Maria Tredicine
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Francesco Ria
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Department Laboratory and Infectious diseases Sciences, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Noemi Poerio
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Matteo Lucchini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Assunta Bianco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Federica De Santis
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Mariagrazia Valentini
- Section of Pathology, Department of Woman, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Valeria De Arcangelis
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Mario Rende
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Anna Maria Stabile
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Alessandra Pistilli
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Chiara Camponeschi
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Institute of Chemical Sciences and Technologies ''Giulio Natta'' (SCITEC)-CNR, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Viviana Nociti
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Massimiliano Mirabella
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Maurizio Fraziano
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Gabriele Di Sante
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| |
Collapse
|
49
|
Verreycken J, Baeten P, Broux B. Regulatory T cell therapy for multiple sclerosis: Breaching (blood-brain) barriers. Hum Vaccin Immunother 2022; 18:2153534. [PMID: 36576251 PMCID: PMC9891682 DOI: 10.1080/21645515.2022.2153534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder causing demyelination and neurodegeneration in the central nervous system. MS is characterized by disturbed motor performance and cognitive impairment. Current MS treatments delay disease progression and reduce relapse rates with general immunomodulation, yet curative therapies are still lacking. Regulatory T cells (Tregs) are able to suppress autoreactive immune cells, which drive MS pathology. However, Tregs are functionally impaired in people with MS. Interestingly, Tregs were recently reported to also have regenerative capacity. Therefore, experts agree that Treg cell therapy has the potential to ameliorate the disease. However, to perform their local anti-inflammatory and regenerative functions in the brain, they must first migrate across the blood-brain barrier (BBB). This review summarizes the reported results concerning the migration of Tregs across the BBB and the influence of Tregs on migration of other immune subsets. Finally, their therapeutic potential is discussed in the context of MS.
Collapse
Affiliation(s)
- Janne Verreycken
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Paulien Baeten
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Bieke Broux
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium,CONTACT Bieke Broux Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Martelarenlaan 42, Hasselt 3500, Belgium
| |
Collapse
|
50
|
Nishihara H, Perriot S, Gastfriend BD, Steinfort M, Cibien C, Soldati S, Matsuo K, Guimbal S, Mathias A, Palecek SP, Shusta EV, Pasquier RD, Engelhardt B. Intrinsic blood-brain barrier dysfunction contributes to multiple sclerosis pathogenesis. Brain 2022; 145:4334-4348. [PMID: 35085379 PMCID: PMC10200307 DOI: 10.1093/brain/awac019] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 07/20/2023] Open
Abstract
Blood-brain barrier (BBB) breakdown and immune cell infiltration into the CNS are early hallmarks of multiple sclerosis (MS). The mechanisms leading to BBB dysfunction are incompletely understood and generally thought to be a consequence of neuroinflammation. Here, we have challenged this view and asked if intrinsic alterations in the BBB of MS patients contribute to MS pathogenesis. To this end, we made use of human induced pluripotent stem cells derived from healthy controls and MS patients and differentiated them into brain microvascular endothelial cell (BMEC)-like cells as in vitro model of the BBB. MS-derived BMEC-like cells showed impaired junctional integrity, barrier properties and efflux pump activity when compared to healthy controls. Also, MS-derived BMEC-like cells displayed an inflammatory phenotype with increased adhesion molecule expression and immune cell interactions. Activation of Wnt/β-catenin signalling in MS-derived endothelial progenitor cells enhanced barrier characteristics and reduced the inflammatory phenotype. Our study provides evidence for an intrinsic impairment of BBB function in MS patients that can be modelled in vitro. Human iPSC-derived BMEC-like cells are thus suitable to explore the molecular underpinnings of BBB dysfunction in MS and will assist in the identification of potential novel therapeutic targets for BBB stabilization.
Collapse
Affiliation(s)
- Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Sylvain Perriot
- Laboratory of Neuroimmunology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Marel Steinfort
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Celine Cibien
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Kinya Matsuo
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Sarah Guimbal
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|