1
|
Zhang G, Yao Q, Long C, Yi P, Song J, Wu L, Wan W, Rao X, Lin Y, Wei G, Ying J, Hua F. Infiltration by monocytes of the central nervous system and its role in multiple sclerosis: reflections on therapeutic strategies. Neural Regen Res 2025; 20:779-793. [PMID: 38886942 PMCID: PMC11433895 DOI: 10.4103/nrr.nrr-d-23-01508] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 02/18/2024] [Indexed: 06/20/2024] Open
Abstract
Mononuclear macrophage infiltration in the central nervous system is a prominent feature of neuroinflammation. Recent studies on the pathogenesis and progression of multiple sclerosis have highlighted the multiple roles of mononuclear macrophages in the neuroinflammatory process. Monocytes play a significant role in neuroinflammation, and managing neuroinflammation by manipulating peripheral monocytes stands out as an effective strategy for the treatment of multiple sclerosis, leading to improved patient outcomes. This review outlines the steps involved in the entry of myeloid monocytes into the central nervous system that are targets for effective intervention: the activation of bone marrow hematopoiesis, migration of monocytes in the blood, and penetration of the blood-brain barrier by monocytes. Finally, we summarize the different monocyte subpopulations and their effects on the central nervous system based on phenotypic differences. As activated microglia resemble monocyte-derived macrophages, it is important to accurately identify the role of monocyte-derived macrophages in disease. Depending on the roles played by monocyte-derived macrophages at different stages of the disease, several of these processes can be interrupted to limit neuroinflammation and improve patient prognosis. Here, we discuss possible strategies to target monocytes in neurological diseases, focusing on three key aspects of monocyte infiltration into the central nervous system, to provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangyong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qing Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Chubing Long
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Ransohoff RM. Selected Aspects of the Neuroimmunology of Cell Therapies for Neurologic Disease: Perspective. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200352. [PMID: 39671535 DOI: 10.1212/nxi.0000000000200352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 12/15/2024]
Abstract
Neurologic disease remains a cause of incalculable suffering, a formidable public health burden, and a wilderness of complex biology and medicine. At the same time, advances in basic science, technology, and the clinical development toolkit bring meaningful benefit for patients along with realistic hope for those whose conditions remain inadequately treated. This perspective focuses on cell-based therapies for neurologic disease, with particular emphasis on neuroimmunologic disorders and on the immunologic considerations of cell therapy for nonimmune conditions. I will consider the use of chimeric antigen receptor (CAR)-T effector cells and regulatory T-cell therapies for autoimmune conditions. I will briefly discuss the immune aspects of pluripotent stem cell (PSC)-derived neuronal therapies. With apologies for the omission, we do not discuss mesenchymal stem cells, glial progenitor cells, or CAR-NK cells, primarily for space limitations.
Collapse
|
3
|
Chen Y, Man-Tak Chu J, Liu JX, Duan YJ, Liang ZK, Zou X, Wei M, Xin WJ, Xu T, Tin-Chun Wong G, Feng X. Double negative T cells promote surgery-induced neuroinflammation, microglial engulfment and cognitive dysfunction via the IL-17/CEBPβ/C3 pathway in adult mice. Brain Behav Immun 2025; 123:965-981. [PMID: 39491565 DOI: 10.1016/j.bbi.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/11/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024] Open
Abstract
CD3(+) CD4(-) CD8(-) double negative T cells (DNTs) manifest themselves in autoimmune diseases and associated inflammation. In the central nervous system, the increased presence of DNTs is associated with the progression of neurological conditions and brain injury. Active DNTs that produce IL-17 have been regarded as a pro-inflammatory phenotype. The IL-17 signaling pathway mediates neuroinflammatory responses by inducing glial activation and producing inflammatory factors. Neuroinflammation is considered integral to the pathogenesis of perioperative neurocognitive disorders (PNDs), commonly developed after surgery in susceptible patients. We and others have demonstrated a significant role for complement C3 in surgery-induced neuroinflammation and cognitive impairment but the regulatory mechanisms for this remain unexplored. We hypothesized that surgery induces DNT infiltration into the CNS that in turn upregulates complement C3 expression and this causes changes that contribute to cognitive impairment. Using an adult murine abdominal surgery model, we investigated perioperative changes in cognitive performance, quantifying the presence of T cell subsets and phenotype, IL-17 signaling pathway activation, glial cell activation and C3 expression in the brain. Postoperative IL-17 specific inhibitor GSK2981278 administration or preoperatively conditional CEBPβ knock-down by AAV9 viral vector were then applied to evaluate the effect of inhibiting IL-17 signaling pathway on postoperative C3 expression and cognitive performance. The results showed an increased hippocampus infiltration of DNTs with augmented IL-17 production, along with C3 upregulation and cognitive impairment. Both inhibition of IL-17 or knock-down of CEBPβ significantly suppressed C3 expression, synaptic engulfment by microglia and attenuated cognitive impairment. These findings indicate that DNTs promote postoperative neuroinflammation and cognitive impairment via the IL-17/CEBPβ/C3 pathway and targeting this IL-17 axis could be a potential therapeutic strategy to ameliorate postoperative neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - John Man-Tak Chu
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, 4Th Floor, Block K, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Jia-Xin Liu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Juan Duan
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zheng-Kai Liang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xin Zou
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ming Wei
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen-Jun Xin
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Xu
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Gordon Tin-Chun Wong
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, 4Th Floor, Block K, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China.
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
4
|
Esposito A, Falco F, Scalia G, Gentile L, Spiezia AL, Corsini G, Manganiello R, Eliano M, Lamagna F, Moccia M, Petracca M, Lanzillo R, Brescia Morra V, Carotenuto A. Association between CD20 + T lymphocytes and neuropsychological findings in multiple sclerosis. Eur J Neurol 2025; 32:e16536. [PMID: 39475271 PMCID: PMC11625923 DOI: 10.1111/ene.16536] [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: 07/23/2024] [Revised: 09/17/2024] [Accepted: 10/15/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND AND PURPOSE CD20+ T lymphocytes are a subset of circulating T cells presenting the CD20+ receptor, a molecular marker of B lineage. CD20+ T lymphocytes are thought to play a pivotal role in multiple sclerosis (MS) pathology, especially at progressive stages. We aimed to investigate the correlation between CD20+ T lymphocytes and neuropsychological features (i.e., cognition, depression, anxiety, fatigue, and sleep quality) in MS patients. METHODS We enrolled 90 MS patients. Each patient underwent cognitive assessment (Brief International Cognitive Assessment for Multiple Sclerosis) and psychometric assessment (modified Fatigue Impact Scale, Beck Anxiety Inventory, Beck Depression Inventory, Pittsburgh Sleep Quality Index). Cognitive status was defined through the cerebral functional score. RESULTS Forty-four of 90 patients were relapsing-remitting (49%) and 46 were progressive patients (51%). Seventy patients (18.9%) showed CD20+ T lymphocytes in peripheral blood with a mean level of 0.38 ± 1.2%. Patients with CD20+ T lymphocytes were more likely to be at progressive phases (76.5% vs. 23.5%, p = 0.02) and showed a higher Expanded Disability Status Scale score (median [range] = 6.0 [1.5-7.5] vs. 3.5 [1-7.5], p = 0.001). Moreover, patients with CD20+ T lymphocytes showed worse cognitive functioning (p = 0.004), higher global fatigue symptoms (p = 0.02), higher cognitive fatigue (p = 0.01), higher psychosocial fatigue (p = 0.005), and a trend toward worse sleep quality (p = 0.06). CONCLUSIONS The presence of CD20+ T lymphocytes in the peripheral blood of MS patients was associated with worse neuropsychological functioning and progressive disease stages. Peripheral CD20+ T lymphocytes could potentially serve as markers for both disease progression and development of fatigue in MS patients.
Collapse
Affiliation(s)
- Antonio Esposito
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Fabrizia Falco
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Giulia Scalia
- Laboratorio di Citometria Clinica e Sperimentale CEINGE‐Biotecnologie Avanzate Franco SalvatoreNaplesItaly
| | - Laura Gentile
- Laboratorio di Citometria Clinica e Sperimentale CEINGE‐Biotecnologie Avanzate Franco SalvatoreNaplesItaly
| | - Antonio Luca Spiezia
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Giuseppe Corsini
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Rosa Manganiello
- Laboratorio di Citometria Clinica e Sperimentale CEINGE‐Biotecnologie Avanzate Franco SalvatoreNaplesItaly
| | - Martina Eliano
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
- Department of PsychologyUniversità degli Studi della Campania L. VanvitelliCasertaItaly
| | - Federica Lamagna
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
- Department of PsychologyUniversità degli Studi della Campania L. VanvitelliCasertaItaly
| | - Marcello Moccia
- Department of Molecular Medicine and Medical BiotechnologyFederico II University of NaplesNaplesItaly
| | - Maria Petracca
- Department of Human NeurosciencesSapienza UniversityRomeItaly
| | - Roberta Lanzillo
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Vincenzo Brescia Morra
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| | - Antonio Carotenuto
- Multiple Sclerosis Clinical Care and Research Center, Department of Neuroscience, Reproductive Science, and OdontostomatologyFederico II University of NaplesNaplesItaly
| |
Collapse
|
5
|
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
|
6
|
Freeman SA, Zéphir H. Anti-CD20 monoclonal antibodies in multiple sclerosis: Rethinking the current treatment strategy. Rev Neurol (Paris) 2024; 180:1047-1058. [PMID: 38599976 DOI: 10.1016/j.neurol.2023.12.013] [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] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 04/12/2024]
Abstract
Anti-CD20 monoclonal antibodies are highly-effective B-cell-depleting therapies in multiple sclerosis (MS). These treatments have expanded the arsenal of highly effective disease-modifying therapies, and have changed the landscape in understanding the pathophysiology of MS and the natural course of the disease. Nevertheless, these treatments come at the cost of immunosuppression and risk of serious infections, diminished vaccination response and treatment-related secondary hypogammaglobulinemia. However, the COVID pandemic has given way to a possibility of readapting these therapies, with most notably extended dosing intervals. While these new strategies show efficacy in maintaining inflammatory MS disease control, and although it is tempting to speculate that tailoring CD20 therapies will reduce the negative outcomes of long-term immunosuppression, it is unknown whether they provide meaningful benefit in reducing the risk of treatment-related secondary hypogammaglobulinemia and serious infections. This review highlights the available anti-CD20 therapies that are available for treating MS patients, and sheds light on encouraging data, which propose that tailoring anti-CD20 monoclonal antibodies is the next step in rethinking the current treatment strategy.
Collapse
Affiliation(s)
- S A Freeman
- Department of Neurology, CRC-SEP, CHU of Toulouse, Toulouse, France; University Toulouse III, Inserm UMR1291, CHU Purpan, Toulouse Institute for Infectious and Inflammatory Diseases (INFINITY), 59000 Toulouse, France.
| | - H Zéphir
- Department of Neurology, CRC-SEP, CHU of Lille, Lille, France; University of Lille, Inserm, CHU of Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), U1172, Lille, France
| |
Collapse
|
7
|
Mohammed EMA. Understanding Multiple Sclerosis Pathophysiology and Current Disease-Modifying Therapies: A Review of Unaddressed Aspects. FRONT BIOSCI-LANDMRK 2024; 29:386. [PMID: 39614433 DOI: 10.31083/j.fbl2911386] [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: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) with an unknown etiology and pathophysiology that is not completely understood. Although great strides have been made in developing disease-modifying therapies (DMTs) that have significantly improved the quality of life for MS patients, these treatments do not entirely prevent disease progression or relapse. Identifying the unaddressed pathophysiological aspects of MS and developing targeted therapies to fill in these gaps are essential in providing long-term relief for patients. Recent research has uncovered some aspects of MS that remain outside the scope of available DMTs, and as such, yield only limited benefits. Despite most MS pathophysiology being targeted by DMTs, many patients still experience disease progression or relapse, indicating that a more detailed understanding is necessary. Thus, this literature review seeks to explore the known aspects of MS pathophysiology, identify the gaps in present DMTs, and explain why current treatments cannot entirely arrest MS progression.
Collapse
Affiliation(s)
- Eiman M A Mohammed
- Kuwait Cancer Control Centre, Department of Medical Laboratory, Molecular Genetics Laboratory, Ministry of Health, 13001 Shuwaikh, Kuwait
| |
Collapse
|
8
|
Häusler D, Weber MS. Towards Treating Multiple Sclerosis Progression. Pharmaceuticals (Basel) 2024; 17:1474. [PMID: 39598386 PMCID: PMC11597358 DOI: 10.3390/ph17111474] [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: 10/08/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system (CNS). In most patients, the disease starts with an acute onset followed by a remission phase, subsequent relapses and a later transition to steady chronic progression. In a minority of patients, this progressive phase develops from the beginning. MS relapses are characterized predominantly by the de novo formation of an inflammatory CNS lesion and the infiltration of immune cells, whereas the pathological features of MS progression include slowly expanding lesions, global brain atrophy and an inflammatory response predominantly mediated by macrophages/microglia. Importantly, this CNS-intrinsic pathophysiology appears to initiate early during the relapsing-remitting disease phase, while it turns into the key clinical MS feature in later stages. Currently approved disease-modifying treatments for MS are effective in modulating peripheral immunity by dampening immune cell activity or preventing the migration of immune cells into the CNS, resulting in the prevention of relapses; however, they show limited success in halting MS progression. In this manuscript, we first describe the pathological mechanisms of MS and summarize the approved therapeutics for MS progression. We also review the treatment options for progressive MS (PMS) that are currently under investigation. Finally, we discuss potential targets for novel treatment strategies in PMS.
Collapse
Affiliation(s)
- Darius Häusler
- Institute of Neuropathology, University Medical Centre, 37075 Goettingen, Germany;
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, 37075 Goettingen, Germany
| | - Martin S. Weber
- Institute of Neuropathology, University Medical Centre, 37075 Goettingen, Germany;
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, 37075 Goettingen, Germany
- Department of Neurology, University Medical Centre, 37075 Goettingen, Germany
| |
Collapse
|
9
|
El Mahdaoui S, von Essen MR, Hansen MM, Romme Christensen J, Sellebjerg F, Søndergaard HB. Profiling of B cells and their subsets by whole blood gene expression analysis versus flow cytometry in multiple sclerosis. Mult Scler Relat Disord 2024; 91:105898. [PMID: 39317145 DOI: 10.1016/j.msard.2024.105898] [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: 07/11/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
We investigated if differentially expressed mRNA targets could be used as surrogate markers for circulating B cells and subsets. In paired blood samples from patients with untreated, anti-CD20-treated, fingolimod-treated, and natalizumab-treated multiple sclerosis, whole blood expression of CD19 correlated with B cell counts determined by flow cytometry, ROR1 with transitional B cells, TCL1A and ZNF727 with naïve B cells, NEXMIF with memory B cells and BCMA with plasmablasts. CD19 expression distinguished patients with B cell repletion and may be used as an alternative to flow cytometry, but NEXMIF was unsuitable for memory B cell monitoring in rituximab-treated patients.
Collapse
Affiliation(s)
- Sahla El Mahdaoui
- 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
| | - Marie Mathilde Hansen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Jeppe Romme Christensen
- 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, University of Copenhagen, Copenhagen, Denmark
| | - Helle Bach Søndergaard
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
10
|
Zhuo Z, Zhang N, Ao F, Hua T, Duan Y, Xu X, Weng J, Cao G, Li K, Zhou F, Li H, Li Y, Han X, Haller S, Barkhof F, Hu G, Shi F, Zhang X, Tian D, Liu Y. Spatial structural abnormality maps associated with cognitive and physical performance in relapsing-remitting multiple sclerosis. Eur Radiol 2024:10.1007/s00330-024-11157-w. [PMID: 39470796 DOI: 10.1007/s00330-024-11157-w] [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: 03/14/2024] [Revised: 07/21/2024] [Accepted: 09/02/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVES We aimed to characterize the brain abnormalities that are associated with the cognitive and physical performance of patients with relapsing-remitting multiple sclerosis (RRMS) using a deep learning algorithm. MATERIALS AND METHODS Three-dimensional (3D) nnU-Net was employed to calculate a novel spatial abnormality map by T1-weighted images and 281 RRMS patients (Dataset-1, male/female = 101/180, median age [range] = 35.0 [17.0, 65.0] years) were categorized into subtypes. Comparison of clinical and MRI features between RRMS subtypes was conducted by Kruskal-Wallis test. Kaplan-Meier analysis was conducted to investigate disability progression in RRMS subtypes. Additional validation using two other RRMS datasets (Dataset-2, n = 33 and Dataset-3, n = 56) was conducted. RESULTS Five RRMS subtypes were identified: (1) a Frontal-I subtype showing preserved cognitive performance and mild physical disability, and low risk of disability worsening; (2) a Frontal-II subtype showing low cognitive scores and severe physical disability with significant brain volume loss, and a high propensity for disability worsening; (3) a temporal-cerebellar subtype demonstrating lowest cognitive scores and severest physical disability among all subtypes but remaining relatively stable during follow-up; (4) an occipital subtype demonstrating similar clinical and imaging characteristics as the Frontal-II subtype, except a large number of relapses at baseline and preserved cognitive performance; and (5) a subcortical subtype showing preserved cognitive performance and low physical disability but a similar prognosis as the occipital and Frontal-II subtypes. Additional validation confirmed the above findings. CONCLUSION Spatial abnormality maps can explain heterogeneity in cognitive and physical performance in RRMS and may contribute to stratified management. KEY POINTS Question Can a deep learning algorithm characterize the brain abnormalities associated with the cognitive and physical performance of patients with RRMS? Findings Five RRMS subtypes were identified by the algorithm that demonstrated variable cognitive and physical performance. Clinical relevance The spatial abnormality maps derived RRMS subtypes had distinct cognitive and physical performances, which have a potential for individually tailored management.
Collapse
Affiliation(s)
- Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Ao
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Radiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Tiantian Hua
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaolu Xu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyuan Weng
- Department of Medical Imaging Product, Neusoft Group Ltd., Shenyang, People's Republic of China
| | - Guanmei Cao
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kuncheng Li
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Haiqing Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Han
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sven Haller
- Department of Imaging and Medical Informatics, University Hospitals of Geneva and Faculty of Medicine of the University of Geneva, Geneva, Switzerland
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London, UK
| | - Geli Hu
- Clinical and Technical Support, Philips Healthcare, Beijing, China
| | - Fudong Shi
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinghu Zhang
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Decai Tian
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Marantos T, Kyriazopoulou E, Angelakis E, Kitsos D, Chondrogianni M, Mpizta G, Papadopoulos A, Giannopoulos S, Voumvourakis K, Tsiodras S. Immunogenicity of a seasonal influenza and a pneumococcal polysaccharide vaccine in multiple sclerosis patients under disease modifying therapies: A single-center prospective study. Vaccine 2024; 42:126001. [PMID: 38796324 DOI: 10.1016/j.vaccine.2024.05.049] [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: 07/09/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Immunogenicity of influenza and pneumococcal vaccines varies and requires further elucidation in patients with multiple sclerosis (MS) under treatment with disease-modifying therapies (DMTs). METHODS Adult MS patients who consented with vaccination after standard-of-care consultation by their treating physicians were enrolled. All received a single dose of an inactivated quadrivalent influenza vaccine and of the 23-valent pneumococcal vaccine. A blood sample was collected before and after four weeks of vaccination for measurement of antibodies against Influenza A, B and S. pneumoniae. Patients were followed-up for adverse events and MS relapse for 12 months. RESULTS One hundred and seventy-two patients (65.7 % female, mean age 42 ± 13 years old, mean MS duration 7.6 ± 7.2 years, 81.4 % under DMTs) were enrolled from November 2019 to March 2020. Antibody measurements were available for 151 patients. Seropositivity for anti-PPSV23 did not differ between baseline and at 4 weeks of follow-up (n = 56, 37.1 %). There was a significant increase of absolute antibody titers post-vaccination for both influenza A and B (p < 0.001). For Influenza A, seropositivity was evident for 57 (37.7 %) patients at 4 weeks compared to 19 (12.6 %) patients at baseline (pMcNemar < 0.001). For Influenza Β, 110 (72.8 %) seroconverted 4 weeks after vaccination compared to 12 (7.9 %) at baseline (pMcNemar < 0.001). Interferon and fumarate did not affect influenza seroconversion while rituximab was associated with lower titers. Mild local AEs (pain, edema) were observed in 23.8 %; no severe AE was reported. Thirty-four patients (19.8 %) had a relapse during the 12-month follow-up; none was attributed to the vaccination. CONCLUSIONS Seroconversion in MS patients on treatment was more frequent following influenza compared to PPSV23 vaccination. Rituximab had an effect on the height of the immune response. Better immunization coverage as well as future evaluation of the breadth of immune response elicited by immunization is necessary for these patients.
Collapse
Affiliation(s)
- Theodoros Marantos
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evdoxia Kyriazopoulou
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Emmanouil Angelakis
- Diagnostic Department and Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
| | - Dimitrios Kitsos
- 2(nd) Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Chondrogianni
- 2(nd) Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Giota Mpizta
- Diagnostic Department and Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
| | - Antonios Papadopoulos
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios Giannopoulos
- 2(nd) Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Sotirios Tsiodras
- 4(th) Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
12
|
Shi Y, Ma J, Li S, Liu C, Liu Y, Chen J, Liu N, Liu S, Huang H. Sex difference in human diseases: mechanistic insights and clinical implications. Signal Transduct Target Ther 2024; 9:238. [PMID: 39256355 PMCID: PMC11387494 DOI: 10.1038/s41392-024-01929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024] Open
Abstract
Sex characteristics exhibit significant disparities in various human diseases, including prevalent cardiovascular diseases, cancers, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Risk profiles and pathological manifestations of these diseases exhibit notable variations between sexes. The underlying reasons for these sex disparities encompass multifactorial elements, such as physiology, genetics, and environment. Recent studies have shown that human body systems demonstrate sex-specific gene expression during critical developmental stages and gene editing processes. These genes, differentially expressed based on different sex, may be regulated by androgen or estrogen-responsive elements, thereby influencing the incidence and presentation of cardiovascular, oncological, metabolic, immune, and neurological diseases across sexes. However, despite the existence of sex differences in patients with human diseases, treatment guidelines predominantly rely on male data due to the underrepresentation of women in clinical trials. At present, there exists a substantial knowledge gap concerning sex-specific mechanisms and clinical treatments for diverse diseases. Therefore, this review aims to elucidate the advances of sex differences on human diseases by examining epidemiological factors, pathogenesis, and innovative progress of clinical treatments in accordance with the distinctive risk characteristics of each disease and provide a new theoretical and practical basis for further optimizing individualized treatment and improving patient prognosis.
Collapse
Affiliation(s)
- Yuncong Shi
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jianshuai Ma
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Sijin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Chao Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yuning Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jie Chen
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ningning Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China.
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Lewis JE, McDaniel HR, Woolger JM, Anzola E, Kraft G. The Characterization of the Th1/Th2 Ratio in Multiple Sclerosis Patients and its Response to a Dietary Supplement Regimen. J Diet Suppl 2024; 21:771-790. [PMID: 39140744 DOI: 10.1080/19390211.2024.2386259] [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] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a debilitating neurodegenerative disease affecting the central nervous system, causing disability and life-threatening complications. The interplay between immune cells and signaling pathways is a topic for investigating novel therapies. Past research has shown how the Th1/Th2 ratio plays a key role in the pathogenesis of MS lesions. Modulating the Th1/Th2 ratios with an efficacious dietary supplement may improve some of the consequences of MS. METHODS Participants (n = 15) diagnosed with MS for an average of 12.4 years (standard deviation = 7.4; range = 2, 25) were enrolled in a clinical trial in which they consumed a dietary supplement regimen daily for 12 months. Venous blood was drawn at baseline and 12-month follow-up and peripheral blood mononuclear cells, cytokines, and growth factors were quantified. Infections, physical functioning, and quality of life were also assessed at baseline and 12 months. RESULTS The IL-2/IL-10 and IFN-γ/IL-10 ratios were significantly higher than those of the healthy adults, and while only IFN-γ/IL-10 increased significantly at 12 months, all ratios other than IFN-γ/TNF-α increased over the course of the intervention. The decrease in yeast infections was inversely correlated with IL-2/TNF-α and IFN-γ/TNF-α. Significant improvements in physical functioning and quality of life correlated with changes in the Th1/Th2 ratios in response to the dietary supplement regimen. CONCLUSIONS The results show that dietary supplementation somewhat impacted the Th1/Th2 ratios over the course of the intervention (toward more Th1 dominance), and those changes were related to various clinical improvements of the participants' symptoms in cognitive, motor, and psychosocial dimensions.
Collapse
Affiliation(s)
| | | | | | - Enrique Anzola
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Garrett Kraft
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
14
|
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
|
15
|
Song Z, Li J, He Y, Wang X, Tian J, Wu Y. A novel PROTAC molecule dBET1 alleviates pathogenesis of experimental autoimmune encephalomyelitis in mice by degrading BRD4. Int Immunopharmacol 2024; 139:112646. [PMID: 39002520 DOI: 10.1016/j.intimp.2024.112646] [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: 01/21/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Neuroinflammation and neurodegeneration are hallmarks of multiple sclerosis (MS). Bromodomain-containing protein 4 (BRD4), a bromodomain and extra-terminal domain (BET) protein family member, is indispensable for the transcription of pro-inflammatory genes. Therefore, inhibiting BRD4 may be a prospective therapeutic approach for modulating the inflammatory response and regulating the course of MS. dBET1, a newly synthesized proteolysis-targeting chimera (PROTAC), exhibits effectively degrades of BRD4. However, the precise effects of dBET1 on MS require further investigation. Therefore, we assessed the effect of dBET1 in experimental autoimmune encephalomyelitis (EAE), a typical MS experimental model. Our findings revealed that BRD4 is mainly expressed in astrocytes and neurons of the spinal cords, and is up-regulated in the spinal cords of EAE mice. The dBET1 attenuated lipopolysaccharide-induced expression of astrocytic pro-inflammatory mediators and inhibited deleterious molecular activity in astrocytes. Correspondingly, dBET1, used in preventive and therapeutic settings, alleviated the behavioral symptoms in EAE mice, as demonstrated by decreased demyelination, alleviated leukocyte infiltration, reduced microglial and astrocyte activation, and diminished inflammatory mediator levels. In addition, dBET1 corrected the imbalance in peripheral T cells and protected blood-brain barrier integrity in EAE mice. The underlying mechanism involved suppressing the phosphoinositide-3-kinase/protein kinase B, mitogen-activated protein kinase /extracellular signal-regulated kinase, and nuclear factor kappa B pathways. In summary, our data strongly suggests that dBET1 is a promising treatment option for MS.
Collapse
Affiliation(s)
- Ziwei Song
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150001, China
| | - Jin Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yijie He
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xiaokun Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jianan Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yun Wu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
16
|
Rabaneda-Lombarte N, Teniente-Serra A, Massuet-Vilamajó A, Ramo-Tello C, Presas-Rodríguez S. Case report: tumefactive demyelinating lesions after the second cycle of alemtuzumab in multiple sclerosis; immune cell profile and biomarkers. Front Immunol 2024; 15:1395749. [PMID: 39021563 PMCID: PMC11251942 DOI: 10.3389/fimmu.2024.1395749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 07/20/2024] Open
Abstract
Objective We present a case of multiple tumefactive demyelinating lesions (TDLs) emerging 24 months after the second cycle of alemtuzumab treatment. Methods A woman with relapsing-remitting multiple sclerosis (MS) discontinued fingolimod treatment due to gestational desire, which resulted in a severe disease exacerbation. Alemtuzumab was initiated, accompanied by regular clinical, radiological, and immunological monitoring. Results She relapsed prior to the second cycle, exhibiting 12 T1Gd+ lesions, and peripheral blood showed an increase in B-cells and a decrease in T-cells. At 24 months following the second cycle, she developed cognitive impairment and multiple T1Gd+ lesions, including TDLs, were evident on the brain MRI. We found not only an increase in B-cells but also in Th1 central memory cells. Th1/Th17 cells increased 3 months before the detection of TDLs. Conclusions TDLs can appear 24 months after the second cycle of alemtuzumab treatment in MS. The increase in Th1/Th17 cells could be a candidate biomarker for TDLs in alemtuzumab-treated MS patients.
Collapse
Affiliation(s)
| | - Aina Teniente-Serra
- Immunology Division, Clinical Laboratory MetroNord (LCMN), Hospital Universitari Germans Trias i Pujol and Research Institute (IGTP), Badalona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Badalona, Spain
| | | | - Cristina Ramo-Tello
- Neurosciences Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | |
Collapse
|
17
|
Hernandez J. Multiple sclerosis treatment review for primary care providers. Nurse Pract 2024; 49:38-47. [PMID: 38915149 PMCID: PMC11186711 DOI: 10.1097/01.npr.0000000000000202] [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] [Indexed: 06/26/2024]
Abstract
ABSTRACT The treatment landscape for multiple sclerosis has dramatically grown in terms of available options and complexity. The various mechanisms of action and safety profiles of these new treatments necessitate that primary care providers remain current in knowledge and practice to provide high-quality care.
Collapse
Affiliation(s)
- Jeffrey Hernandez
- Jeffrey Hernandez is an MS-certified NP and supervisor of advanced practice providers at the MS Center at University of Miami in Miami, Fla
| |
Collapse
|
18
|
Spiezia AL, Scalia G, Petracca M, Caliendo D, Moccia M, Fiore A, Cerbone V, Lanzillo R, Brescia Morra V, Carotenuto A. Effect of siponimod on lymphocyte subsets in active secondary progressive multiple sclerosis and clinical implications. J Neurol 2024; 271:4281-4291. [PMID: 38632126 PMCID: PMC11233419 DOI: 10.1007/s00415-024-12362-9] [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: 01/04/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Circulating immune cells play a pathogenic role in multiple sclerosis (MS). However, the role of specific lymphocyte subpopulations is not unveiled yet, especially in progressive stages. We aimed to investigate lymphocyte changes during siponimod treatment in active secondary progressive MS (aSPMS) and their associations with clinical outcomes. METHODS We enrolled 46 aSPMS patients starting on siponimod treatment with at least 6 months of follow-up and two visits within the scheduled timeframes and 14 sex- and age-matched healthy controls (HCs). Clinical and laboratory data were collected retrospectively at baseline, 3rd, 6th, 12th, and 24th month for MS patients, and at baseline for HCs. RESULTS At baseline SPMS patients presented with increased naïve regulatory T lymphocytes (p = 0.02) vs. HCs. Over time, SPMS patients showed decreased T CD4+ (coeff. range = -24/-17, 95% CI range = -31.60 to -10.40), B lymphocyte (coeff. range = -3.77/-2.54, 95% CI range = -6.02 to -0.35), memory regulatory B cells (coeff. range = -0.78/-0.57, 95% CI range = -1.24 to -0.17) and CD4/CD8 ratio (coeff. range = -4.44/-0.67, 95% CI range = -1.61 to -0.17) from month 3 thereafter vs. baseline, and reduced CD3+CD20+ lymphocytes from month 12 thereafter (coeff. range = -0.32/-0.24, 95% CI range = -0.59 to -0.03). Patients not experiencing disability progression while on siponimod treatment showed B lymphocyte reduction from month 3 (coeff. range = -4.23/-2.32, 95% CI range = -7.53 to -0.15) and CD3+CD20+ lymphocyte reduction from month 12 (coeff. range = -0.32/-0.24, 95% CI range = -0.59 to -0.03) vs. patients experiencing progression. CONCLUSIONS Patients treated with siponimod showed a T and B lymphocyte reduction, especially CD4+, CD3+CD20+ and naïve regulatory T cells and memory regulatory B cells. Disability progression while on siponimod treatment was associated with a less pronounced effect on B and CD3+CD20+ lymphocytes.
Collapse
Affiliation(s)
- Antonio Luca Spiezia
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Giulia Scalia
- Clinical and Experimental Cytometry Unit, Centre for Advanced Biotechnology Franco Salvatore, CEINGE, Naples, Italy
| | - Maria Petracca
- Department of Human Neurosciences, Sapienza University, Rome, Italy
| | - Daniele Caliendo
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Marcello Moccia
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Antonia Fiore
- Clinical and Experimental Cytometry Unit, Centre for Advanced Biotechnology Franco Salvatore, CEINGE, Naples, Italy
| | - Vincenza Cerbone
- Clinical and Experimental Cytometry Unit, Centre for Advanced Biotechnology Franco Salvatore, CEINGE, Naples, Italy
| | - Roberta Lanzillo
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Vincenzo Brescia Morra
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Antonio Carotenuto
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
19
|
Tatomir A, Anselmo F, Boodhoo D, Chen H, Mekala AP, Nguyen V, Cuevas J, Rus V, Rus H. Multiple sclerosis disease activity, a multi-biomarker score of disease activity and response to treatment in multiple sclerosis. Front Immunol 2024; 15:1338585. [PMID: 38994359 PMCID: PMC11236682 DOI: 10.3389/fimmu.2024.1338585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Regular assessment of disease activity in relapsing-remitting multiple sclerosis (RRMS) is required to optimize clinical outcomes. Biomarkers can be a valuable tool for measuring disease activity in multiple sclerosis (MS) if they reflect the pathological processes underlying MS pathogenicity. In this pilot study, we combined multiple biomarkers previously analyzed in RRMS patients into an MS disease activity (MSDA) score to evaluate their ability to predict relapses and treatment response to glatiramer acetate (GA). Response Gene to Complement 32 (RGC-32), FasL, IL-21, SIRT1, phosphorylated SIRT1 (p-SIRT1), and JNK1 p54 levels were used to generate cut-off values for each biomarker. Any value below the cutoff for RGC-32, FasL SIRT1, or p-SIRT1 or above the cutoff for IL-21 or JNK1 p54 was given a +1 value, indicating relapse or lack of response to GA. Any value above the cutoff value for RGC-32, FasL, SIRT1, p-SIRT1 or below that for IL-21 or JNK1 p54 was given a -1 value, indicating clinical stability or response to GA. An MSDA score above +1 indicated a relapse or lack of response to treatment. An MSDA score below -1 indicated clinical stability or response to treatment. Our results showed that the MSDA scores generated using either four or six biomarkers had a higher sensitivity and specificity and significantly correlated with the expanded disability status scale. Although these results suggest that the MSDA test can be useful for monitoring therapeutic response to biologic agents and assessing clinically challenging situations, the present findings need to be confirmed in larger studies.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| | - Freidrich Anselmo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Hegang Chen
- Department of Epidemiology and Public Health, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam P. Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jacob Cuevas
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| |
Collapse
|
20
|
Yang Y, Bai Q, Liu F, Zhang S, Tang W, Liu L, Xing Z, Wang H, Zhang C, Yang Y, Fan H. Establishment of the Diagnostic Signature of Ferroptosis Genes in Multiple Sclerosis. Biochem Genet 2024:10.1007/s10528-024-10832-3. [PMID: 38886317 DOI: 10.1007/s10528-024-10832-3] [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: 01/17/2024] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Ferroptosis is a novel form of membrane-dependent cell death that differs from other cell death modalities such as necrosis, apoptosis, and autophagy. Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system primarily affecting brain and spinal cord neurons. Although the pathogenesis of these two conditions may seem unrelated, recent studies have indicated a connection between ferroptosis and multiple sclerosis. In fact, ferroptosis plays a significant role in the development of MS, as evidenced by the presence of elevated iron levels and iron metabolism abnormalities in the brains, spinal cords, and other neurons of MS patients. These abnormalities disrupt iron homeostasis within cells, leading to the occurrence of ferroptosis. However, there is currently a lack of research on the diagnostic value of ferroptosis-related genes in multiple sclerosis. In this study, we employed bioinformatics methods to identify ferroptosis-related genes (ATM, GSK3B, HMGCR, KLF2, MAPK1, NFE2L1, NRAS, PCBP1, PIK3CA, RPL8, VDAC3) associated with the diagnosis of multiple sclerosis and constructed a diagnostic model. The results demonstrated that the diagnostic model accurately identified the patients' condition. Subsequently, subgroup analysis was performed based on the expression levels of ferroptosis-related genes, dividing patients into high and low expression groups. The results showed differences in immune function and immune cell infiltration between the two groups. Our study not only confirms the correlation between ferroptosis and multiple sclerosis but also demonstrates the diagnostic value of ferroptosis-related genes in the disease. This provides guidance for clinical practice and direction for further mechanistic research.
Collapse
Affiliation(s)
- Yang Yang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qianqian Bai
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Fangfei Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Shumin Zhang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Wenchao Tang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Ling Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Zhehua Xing
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Hao Wang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Chi Zhang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Yanhui Yang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
21
|
Liu J, Liu F, Liang T, Zhou Y, Su X, Li X, Zeng J, Qu P, Wang Y, Chen F, Lei Q, Li G, Cheng P. The roles of Th cells in myocardial infarction. Cell Death Discov 2024; 10:287. [PMID: 38879568 PMCID: PMC11180143 DOI: 10.1038/s41420-024-02064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Myocardial infarction, commonly known as a heart attack, is a serious condition caused by the abrupt stoppage of blood flow to a part of the heart, leading to tissue damage. A significant aspect of this condition is reperfusion injury, which occurs when blood flow is restored but exacerbates the damage. This review first addresses the role of the innate immune system, including neutrophils and macrophages, in the cascade of events leading to myocardial infarction and reperfusion injury. It then shifts focus to the critical involvement of CD4+ T helper cells in these processes. These cells, pivotal in regulating the immune response and tissue recovery, include various subpopulations such as Th1, Th2, Th9, Th17, and Th22, each playing a unique role in the pathophysiology of myocardial infarction and reperfusion injury. These subpopulations contribute to the injury process through diverse mechanisms, with cytokines such as IFN-γ and IL-4 influencing the balance between tissue repair and injury exacerbation. Understanding the interplay between the innate immune system and CD4+ T helper cells, along with their cytokines, is crucial for developing targeted therapies to mitigate myocardial infarction and reperfusion injury, ultimately improving outcomes for cardiac patients.
Collapse
Affiliation(s)
- Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiaohan Su
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xue Li
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jiao Zeng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Fuli Chen
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Lei
- Department of Anesthesiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
22
|
Haghikia A, Schett G, Mougiakakos D. B cell-targeting chimeric antigen receptor T cells as an emerging therapy in neuroimmunological diseases. Lancet Neurol 2024; 23:615-624. [PMID: 38760099 DOI: 10.1016/s1474-4422(24)00140-6] [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: 12/11/2023] [Revised: 03/07/2024] [Accepted: 03/27/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Neuroimmunology research and development has been marked by substantial advances, particularly in the treatment of neuroimmunological diseases, such as multiple sclerosis, myasthenia gravis, neuromyelitis optica spectrum disorders, and myelin oligodendrocyte glycoprotein antibody disease. With more than 20 drugs approved for multiple sclerosis alone, treatment has become more personalised. The approval of disease-modifying therapies, particularly those targeting B cells, has highlighted the role of immunotherapeutic interventions in the management of these diseases. Despite these successes, challenges remain, particularly for patients who do not respond to conventional therapies, underscoring the need for innovative approaches. RECENT DEVELOPMENTS The approval of monoclonal antibodies, such as ocrelizumab and ofatumumab, which target CD20, and inebilizumab, which targets CD19, for the treatment of various neuroimmunological diseases reflects progress in the understanding and management of B-cell activity. However, the limitations of these therapies in halting disease progression or activity in patients with multiple sclerosis or neuromyelitis optica spectrum disorders have prompted the exploration of cell-based therapies, particularly chimeric antigen receptor (CAR) T cells. Initially successful in the treatment of B cell-derived malignancies, CAR T cells offer a novel therapeutic mechanism by directly targeting and eliminating B cells, potentially overcoming the shortcomings of antibody-mediated B cell depletion. WHERE NEXT?: The use of CAR T cells in autoimmune diseases and B cell-driven neuroimmunological diseases shows promise as a targeted and durable option. CAR T cells act autonomously, penetrating deep tissue and effectively depleting B cells, especially in the CNS. Although the therapeutic potential of CAR T cells is substantial, their application faces hurdles such as complex logistics and management of therapy-associated toxic effects. Ongoing and upcoming clinical trials will be crucial in determining the safety, efficacy, and applicability of CAR T cells. As research progresses, CAR T cell therapy has the potential to transform treatment for patients with neuroimmunological diseases. It could offer extended periods of remission and a new standard in the management of autoimmune and neuroimmunological disorders.
Collapse
Affiliation(s)
- Aiden Haghikia
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich Alexander Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Haematology, Oncology, and Cell Therapy and Oncology and Health Campus Immunology, Infectiology, and Inflammation (GCI(3)), Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
23
|
Li R, Lei Y, Rezk A, Diego A Espinoza, Wang J, Feng H, Zhang B, Barcelos IP, Zhang H, Yu J, Huo X, Zhu F, Yang C, Tang H, Goldstein AC, Banwell BL, Hakonarson H, Xu H, Mingueneau M, Sun B, Li H, Bar-Or A. Oxidative phosphorylation regulates B cell effector cytokines and promotes inflammation in multiple sclerosis. Sci Immunol 2024; 9:eadk0865. [PMID: 38701189 DOI: 10.1126/sciimmunol.adk0865] [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: 08/02/2023] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
Dysregulated B cell cytokine production contributes to pathogenesis of immune-mediated diseases including multiple sclerosis (MS); however, the underlying mechanisms are poorly understood. In this study we investigated how cytokine secretion by pro-inflammatory (GM-CSF-expressing) and anti-inflammatory (IL-10-expressing) B cells is regulated. Pro-inflammatory human B cells required increased oxidative phosphorylation (OXPHOS) compared with anti-inflammatory B cells. OXPHOS reciprocally modulated pro- and anti-inflammatory B cell cytokines through regulation of adenosine triphosphate (ATP) signaling. Partial inhibition of OXPHOS or ATP-signaling including with BTK inhibition resulted in an anti-inflammatory B cell cytokine shift, reversed the B cell cytokine imbalance in patients with MS, and ameliorated neuroinflammation in a myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalitis mouse model. Our study identifies how pro- and anti-inflammatory cytokines are metabolically regulated in B cells and identifies ATP and its metabolites as a "fourth signal" that shapes B cell responses and is a potential target for restoring the B cell cytokine balance in autoimmune diseases.
Collapse
Affiliation(s)
- Rui Li
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Yanting Lei
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ayman Rezk
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diego A Espinoza
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jing Wang
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Huiru Feng
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Bo Zhang
- Institute of Immunotherapy and Department of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Isabella P Barcelos
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hang Zhang
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Jing Yu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xinrui Huo
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Fangyi Zhu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Changxin Yang
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hao Tang
- MS Research Unit, Biogen, Cambridge, MA 02142, USA
| | - Amy C Goldstein
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brenda L Banwell
- Division of Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Hongwei Xu
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | | | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- MS Research Unit, Biogen, Cambridge, MA 02142, USA
| |
Collapse
|
24
|
Carnero Contentti E, Rojas JI, Giachello S, Henestroza P, Lopez PA. Smoking and Health-Related Quality of Life in Patients With Multiple Sclerosis From Latin America. Int J MS Care 2024; 26:187-193. [PMID: 39072226 PMCID: PMC11273276 DOI: 10.7224/1537-2073.2023-053] [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: 07/30/2024]
Abstract
BACKGROUND Tobacco smoking is an important, modifiable, environmental risk factor for multiple sclerosis (MS) with a relevant impact on health-related quality of life (HRQOL). We aimed to assess the use of tobacco in individuals with MS from Latin America (LATAM), and its impact on HRQOL. METHODS We conducted a cross-sectional study based on a LATAM web-based survey. Demographics, social and clinical data, information on physical disability, and HRQOL scores were collected using the MS Impact Scale-29 (MSIS-29), the Fatigue Severity Scale (FSS), and the Hospital Anxiety and Depression Scale-Anxiety (HADS-A). Individuals with MS were classified at the time of the survey as follows: never-smokers (ie, patients who reported they had never smoked), past smokers (those who had smoked tobacco but not during the past year), or current smokers. For the analysis, groups were compared. RESULTS 425 patients (74.6% female) from 17 LATAM countries were included, mean age 43.6 ± 11 years and median Expanded Disability Status Scale score 2. There were 122 (28.7%) current smokers, 178 (41.9%) past smokers, and 125 (30.4%) never-smokers. Current smokers had significantly higher MSIS-29 physical (physical worsening), FSS (fatigue), and HADS-A (anxiety) scores compared with past and never-smokers after being adjusted for covariables. No significant differences were observed in any of the other analyzed demographic, clinical, and therapeutic variables. Thirty percent of the current and past smokers groups had never had their neurologists discuss smoking cessation with them. CONCLUSIONS Individuals with MS who were current smokers had higher fatigue and anxiety scores and worse HRQOL compared with past and never-smokers.
Collapse
Affiliation(s)
- Edgar Carnero Contentti
- From the Neuroimmunology Unit, Department of Neuroscience, Hospital Alemán, Buenos Aires, Argentina
| | - Juan I. Rojas
- From the Centro de Esclerosis Múltiple de Buenos Aires, Buenos Aires, Argentina
| | - Susana Giachello
- From the Asociación de Lucha Contra la Esclerosis Múltiple, Buenos Aires, Argentina
| | - Paula Henestroza
- From the Asociación de Lucha Contra la Esclerosis Múltiple, Buenos Aires, Argentina
| | - Pablo A. Lopez
- From the Neuroimmunology Unit, Department of Neuroscience, Hospital Alemán, Buenos Aires, Argentina
| |
Collapse
|
25
|
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
|
26
|
Zhang S, Zhang M, Zhang L, Wang Z, Tang S, Yang X, Li Z, Feng J, Qin X. Identification of Y‒linked biomarkers and exploration of immune infiltration of normal-appearing gray matter in multiple sclerosis by bioinformatic analysis. Heliyon 2024; 10:e28085. [PMID: 38515685 PMCID: PMC10956066 DOI: 10.1016/j.heliyon.2024.e28085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Background The knowledge of normal‒appearing cortical gray matter (NAGM) in multiple sclerosis (MS) remains unclear. In this study, we aimed to identify diagnostic biomarkers and explore the immune infiltration characteristics of NAGM in MS through bioinformatic analysis and validation in vivo. Methods Differentially expressed genes (DEGs) were analyzed. Subsequently, the functional pathways of the DEGs were determined. After screening the overlapping DEGs of MS with two machine learning methods, the biomarkers' efficacy and the expression levels of overlapping DEGs were calculated. Quantitative reverse transcription polymerase chain reaction (qRT‒PCR) identified the robust diagnostic biomarkers. Additionally, infiltrating immune cell populations were estimated and correlated with the biomarkers. Finally, the characteristics of immune infiltration of NAGM from MS were evaluated. Results A total of 98 DEGs were identified. They participated in sensory transduction of the olfactory system, synaptic signaling, and immune responses. Nine overlapping genes were screened by machine learning methods. After verified by ROC curve, four genes, namely HLA‒DRB1, RPS4Y1, EIF1AY and USP9Y, were screened as candidate biomarkers. The mRNA expression of RPS4Y1 and USP9Y was significantly lower in MS patients than that in the controls. They were selected as the robust diagnostic biomarkers for male MS patients. RPS4Y1 and USP9Y were both positively correlated with memory B cells. Moreover, naive CD4+ T cells and monocytes were increased in the NAGM of MS patients compared with those in controls. Conclusions Low expressed Y‒linked genes, RPS4Y1 and USP9Y, were identified as diagnostic biomarkers for MS in male patients. The inhomogeneity of immune cells in NAGM might exacerbate intricate interplay between the CNS and the immune system in the MS.
Collapse
Affiliation(s)
| | | | - Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Zijie Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Shi Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaolin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Zhizhong Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
27
|
Cortes-Figueiredo F, Asseyer S, Chien C, Zimmermann HG, Ruprecht K, Schmitz-Hübsch T, Bellmann-Strobl J, Paul F, Morais VA. CD4 + T cell mitochondrial genotype in Multiple Sclerosis: a cross-sectional and longitudinal analysis. Sci Rep 2024; 14:7507. [PMID: 38553515 PMCID: PMC10980703 DOI: 10.1038/s41598-024-57592-z] [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: 03/29/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system (CNS), with a largely unknown etiology, where mitochondrial dysfunction likely contributes to neuroaxonal loss and brain atrophy. Mirroring the CNS, peripheral immune cells from patients with MS, particularly CD4+ T cells, show inappropriate mitochondrial phenotypes and/or oxidative phosphorylation (OxPhos) insufficiency, with a still unknown contribution of mitochondrial DNA (mtDNA). We hypothesized that mitochondrial genotype in CD4+ T cells might influence MS disease activity and progression. Thus, we performed a retrospective cross-sectional and longitudinal study on patients with a recent diagnosis of either Clinically Isolated Syndrome (CIS) or Relapsing-Remitting MS (RRMS) at two timepoints: 6 months (VIS1) and 36 months (VIS2) after disease onset. Our primary outcomes were the differences in mtDNA extracted from CD4+ T cells between: (I) patients with CIS/RRMS (PwMS) at VIS1 and age- and sex-matched healthy controls (HC), in the cross-sectional analysis, and (II) different diagnostic evolutions in PwMS from VIS1 to VIS2, in the longitudinal analysis. We successfully performed mtDNA whole genome sequencing (mean coverage: 2055.77 reads/base pair) in 183 samples (61 triplets). Nonetheless, mitochondrial genotype was not associated with a diagnosis of CIS/RRMS, nor with longitudinal diagnostic evolution.
Collapse
Affiliation(s)
- Filipe Cortes-Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanna Asseyer
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudia Chien
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hanna G Zimmermann
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Einstein Center Digital Future, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité - Universitätsmedizin Berlin, Berlin, Germany.
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| | - Vanessa A Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
28
|
Cross AH, Gelfand JM, Thebault S, Bennett JL, von Büdingen HC, Cameron B, Carruthers R, Edwards K, Fallis R, Gerstein R, Giacomini PS, Greenberg B, Hafler DA, Ionete C, Kaunzner UW, Kodama L, Lock C, Longbrake EE, Musch B, Pardo G, Piehl F, Weber MS, Yuen S, Ziemssen T, Bose G, Freedman MS, Anania VG, Ramesh A, Winger RC, Jia X, Herman A, Harp C, Bar-Or A. Emerging Cerebrospinal Fluid Biomarkers of Disease Activity and Progression in Multiple Sclerosis. JAMA Neurol 2024:2816158. [PMID: 38466277 DOI: 10.1001/jamaneurol.2024.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Importance Biomarkers distinguishing nonrelapsing progressive disease biology from relapsing biology in multiple sclerosis (MS) are lacking. Cerebrospinal fluid (CSF) is an accessible fluid that most closely reflects central nervous system biology. Objective To identify CSF biological measures associated with progressive MS pathobiology. Design, Setting, and Participants This cohort study assessed data from 2 prospective MS cohorts: a test cohort provided serial CSF, clinical, and imaging assessments in a multicenter study of patients with relapsing MS (RMS) or primary progressive MS (PPMS) who were initiating anti-CD20 treatment (recruitment: 2016-2018; analysis: 2020-2023). A single-site confirmation cohort was used to assess CSF at baseline and long-term (>10 year) clinical follow-up (analysis: 2022-2023). Exposures Test-cohort participants initiated standard-of-care ocrelizumab treatment. Confirmation-cohort participants were untreated or received standard-of-care disease-modifying MS therapies. Main Outcomes and Measures Twenty-five CSF markers, including neurofilament light chain, neurofilament heavy chain, and glial fibrillary acid protein (GFAP); 24-week confirmed disability progression (CDP24); and brain magnetic resonance imaging measures reflecting focal injury, tissue loss, and progressive biology (slowly expanding lesions [SELs]). Results The test cohort (n = 131) included 100 patients with RMS (mean [SD] age, 36.6 [10.4] years; 68 [68%] female and 32 [32%] male; Expanded Disability Status Scale [EDSS] score, 0-5.5), and 31 patients with PPMS (mean [SD] age, 44.9 [7.4] years; 15 [48%] female and 16 [52%] male; EDSS score, 3.0-6.5). The confirmation cohort (n = 68) included 41 patients with RMS and 27 with PPMS enrolled at diagnosis (age, 40 years [range, 20-61 years]; 47 [69%] female and 21 [31%] male). In the test cohort, GFAP was correlated with SEL count (r = 0.33), greater proportion of T2 lesion volume from SELs (r = 0.24), and lower T1-weighted intensity within SELs (r = -0.33) but not with acute inflammatory measures. Neurofilament heavy chain was correlated with SEL count (r = 0.25) and lower T1-weighted intensity within SELs (r = -0.28). Immune markers correlated with measures of acute inflammation and, unlike GFAP, were impacted by anti-CD20. In the confirmation cohort, higher baseline CSF GFAP levels were associated with long-term CDP24 (hazard ratio, 2.1; 95% CI, 1.3-3.4; P = .002). Conclusions and Relevance In this study, activated glial markers (in particular GFAP) and neurofilament heavy chain were associated specifically with nonrelapsing progressive disease outcomes (independent of acute inflammatory activity). Elevated CSF GFAP was associated with long-term MS disease progression.
Collapse
Affiliation(s)
- Anne H Cross
- Washington University School of Medicine, St Louis, Missouri
| | | | - Simon Thebault
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | | | | | | | | | - Robert Fallis
- The Ohio State University Wexner Medical Center, Columbus
| | | | | | | | | | | | | | - Lay Kodama
- Genentech, South San Francisco, California
| | | | | | | | | | | | - Martin S Weber
- Institute of Neuropathology, Department of Neurology, University Medical Center, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| | | | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, Dresden, Germany
| | - Gauruv Bose
- Department of Medicine in Neurology, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Mark S Freedman
- Department of Medicine in Neurology, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | | | | | | - Ann Herman
- Genentech, South San Francisco, California
| | | | - Amit Bar-Or
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
29
|
Yazdanpanah E, Dadfar S, Shadab A, Orooji N, Nemati M, Pazoki A, Esmaeili SA, Baharlou R, Haghmorad D. Berberine: A natural modulator of immune cells in multiple sclerosis. Immun Inflamm Dis 2024; 12:e1213. [PMID: 38477663 DOI: 10.1002/iid3.1213] [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: 08/25/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Berberine is a benzylisoquinoline alkaloid found in such plants as Berberis vulgaris, Berberis aristata, and others, revealing a variety of pharmacological properties as a result of interacting with different cellular and molecular targets. Recent studies have shown the immunomodulatory effects of Berberine which result from its impacts on immune cells and immune response mediators such as diverse T lymphocyte subsets, dendritic cells (DCs), and different inflammatory cytokines. Multiple sclerosis (MS) is a chronic disabling and neurodegenerative disease of the central nervous system (CNS) characterized by the recruitment of autoreactive T cells into the CNS causing demyelination, axonal damage, and oligodendrocyte loss. There have been considerable changes discovered in MS regards to the function and frequency of T cell subsets such as Th1 cells, Th17 cells, Th2 cells, Treg cells, and DCs. In the current research, we reviewed the outcomes of in vitro, experimental, and clinical investigations concerning the modulatory effects that Berberine provides on the function and numbers of T cell subsets and DCs, as well as important cytokines that are involved in MS.
Collapse
Affiliation(s)
- Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepehr Dadfar
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Niloufar Orooji
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - MohammadHossein Nemati
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Alireza Pazoki
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
30
|
Seyedmirzaei H, Salabat D, KamaliZonouzi S, Teixeira AL, Rezaei N. Risk of MS relapse and deterioration after COVID-19: A systematic review and meta-analysis. Mult Scler Relat Disord 2024; 83:105472. [PMID: 38316078 DOI: 10.1016/j.msard.2024.105472] [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: 12/11/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Upper respiratory viral infections have long been considered triggers for multiple sclerosis (MS) relapse and exacerbation. The possible effects of SARS-CoV-2 infection on MS relapse and deterioration remain controversial. METHODS We systematically searched PubMed, Scopus, Embase, Cochrane, and Web of Science databases to find relevant studies assessing changes in relapse rates or Expanded Disability Status Scale (EDSS) following COVID-19 in people with MS. Meta-analyses were performed, and to investigate sources of heterogeneity, subgroup analysis, meta-regression, and sensitivity analysis were conducted. RESULTS We included 14 studies in our systematic review and meta-analysis. The meta-analysis demonstrated that COVID-19 was not associated with a rise in relapse rate (risk ratio (RR): 0.97, 95 % confidence interval (CI): 0.67, 1.41, p-value: 0.87) or a rise in EDSS (standardized mean difference (SMD): -0.09, 95 % CI: -0.22, 0.03, p-value: 0.13). The analysis of EDSS changes indicated a significant heterogeneity (I2: 55 %, p-value: 0.01). Other analyses were not statistically significant. CONCLUSIONS COVID-19 infection was not associated with an increased risk of relapse and clinical deterioration in people with MS.
Collapse
Affiliation(s)
- Homa Seyedmirzaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran; Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | - Dorsa Salabat
- Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sara KamaliZonouzi
- Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Antônio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Perez-Paramo YX, Dufield D, Veeramachaneni R, Parkhurst E, Harp C, Ramesh A, Winger RC, Cross AH, Gelfand JM, Bar-Or A, Mathews WR, Anania VG. Development of an LC-MS/MS Method to Measure Sphingolipids in CSF from Patients with Multiple Sclerosis. Mol Pharmacol 2024; 105:121-130. [PMID: 38182433 DOI: 10.1124/molpharm.123.000779] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Multiple sclerosis is an inflammatory and degenerative disease characterized by different clinical courses including relapsing multiple sclerosis (RMS) and primary progressive multiple sclerosis (PPMS). A hallmark of patients with multiple sclerosis (pwMS) includes a putative autoimmune response, which results in demyelination and neuroaxonal damage in the central nervous system. Sphingolipids in cerebrospinal fluid (CSF) have been proposed as potential biomarkers reflective of disease activity in pwMS. Hence, sensitive methods to accurately quantify sphingolipids in CSF are needed. In this study, we report the development of a sensitive high-throughput multiplexed liquid chromatography coupled to a tandem mass spectrometry method to perform quantitation on 14 species of sphingolipids in human CSF. We applied this method to measure CSF sphingolipids in healthy controls (n = 10), PPMS (n = 27), and RMS (n = 17) patients before and after ocrelizumab treatment. The median CSF levels of the 14 sphingolipids measured herein was higher in PPMS (17.2 ng/mL) and RMS (17.6 ng/mL) when compared with the healthy controls (13.8 ng/mL). Levels of sphingolipids were decreased by 8.6% at week 52 after treatment with ocrelizumab in RMS patients but not in PPMS patients. Specifically, C16 glucosylceramide (-26%; P = 0.004) and C18 ceramides (-13%; P = 0.042) decreased from baseline in RMS patients. Additionally, in PPMS patients C16 glucosylceramide levels correlated with CSF neurofilament heavy levels at baseline (Rho =0.532; P = 0.004) and after treatment (Rho =0.424; P = 0.028). Collectively, these results indicate that CSF sphingolipid levels are altered in pwMS and treatment with ocrelizumab results in significant shifts in the sphingolipid profile that may reflect a reduction in disease activity supporting further investigation into sphingolipids as tools to monitor disease state. SIGNIFICANCE STATEMENT: This study describes the development of a new method to measure 14 sphingolipid species in CSF. These results demonstrate that sphingolipids levels are elevated in CSF from pwMS compared to healthy controls. Distinct sphingolipid signatures were observed between patients with different clinical disease courses, and these lipid signatures changed after treatment with ocrelizumab, especially in RMS patients. This method enables further investigation into the role of sphingolipids as candidate biomarkers in pwMS and other central nervous system disorders.
Collapse
Affiliation(s)
- Yadira X Perez-Paramo
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Dawn Dufield
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Rathna Veeramachaneni
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Emily Parkhurst
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Christopher Harp
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Akshaya Ramesh
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Ryan C Winger
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Anne H Cross
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Jeffrey M Gelfand
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Amit Bar-Or
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - W Rodney Mathews
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| | - Veronica G Anania
- Department of Translational Medicine, Genentech, Inc., South San Francisco, California (Y.X.P.-P., C.H., A.R., R.C.W., W.R.M., V.G.A.); KCAS Bioanalytical Sciences, Olathe, Kansas (D.D., R.V., E.P.); Washington University School of Medicine, St Louis, Missouri (A.H.C.); University of California, San Francisco, California (J.M.G.); and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B.-O.)
| |
Collapse
|
32
|
Ongphichetmetha T, Jitprapaikulsan J, Siritho S, Rattanathamsakul N, Detweeratham T, Prayoonwiwat N. Efficacy and safety of rituximab in multiple sclerosis and neuromyelitis optica spectrum disorder. Sci Rep 2024; 14:3503. [PMID: 38347079 PMCID: PMC10861443 DOI: 10.1038/s41598-024-53838-y] [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: 11/23/2023] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
In Thailand, resource limitations lead many multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) patients to use off-label immunosuppressants. This study assesses the efficacy and safety of rituximab (RTX) with a CD19-based reinfusion regimen among Thai MS and NMOSD patients. A retrospective review of patients at the Faculty of Medicine Siriraj Hospital from January 1994 to April 2023 was conducted. The primary outcome assessed was the change in annualized relapse rate (ARR) for patients using RTX for over a year. Secondary outcomes included changes in the Expanded Disability Status Scale (EDSS) scores, time to the first relapse after RTX initiation for patients using RTX for over a year, and an evaluation of the safety of RTX. The study encompassed 36 MS and 39 NMOSD patients. A majority of patients (91.7% of MS and 79.5% of NMOSD) experienced no relapses during a median follow-up of 30 months (Interquartile range [IQR] 20-46) and 31 months (IQR 23-41), respectively. The median ARR significantly decreased in both MS (from 0.77 [IQR 0.42-1.83] to 0 [IQR 0-0], p < 0.001) and NMOSD (from 0.92 [IQR 0.68-1.78] to 0 [IQR 0-0.17], p < 0.001) patients after switching to RTX, with no difference between those following a fixed 6-month time point regimen and a CD19-based reinfusion regimen. Median EDSS scores improved significantly at the last follow-up visit in both groups. The mean time to the first subsequent relapse was 8.3 ± 3.0 months in MS and 6.8 ± 1.7 months in NMOSD. Mild adverse drug reactions occurred in 44% of patients. RTX effectively prevents relapses in Thai MS and NMOSD patients, with no observed serious adverse drug reactions.
Collapse
Affiliation(s)
- Tatchaporn Ongphichetmetha
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jiraporn Jitprapaikulsan
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sasitorn Siritho
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Bumrungrad International Hospital, Bangkok, 10110, Thailand
| | - Natthapon Rattanathamsakul
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thammachet Detweeratham
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
| | - Naraporn Prayoonwiwat
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand.
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
33
|
He Z, Chen Q, Wang K, Lin J, Peng Y, Zhang J, Yan X, Jie Y. Single-cell transcriptomics analysis of cellular heterogeneity and immune mechanisms in neurodegenerative diseases. Eur J Neurosci 2024; 59:333-357. [PMID: 38221677 DOI: 10.1111/ejn.16242] [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: 05/17/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024]
Abstract
Single-cell transcriptomics analysis is an advanced technology that can describe the intracellular transcriptome in complex tissues. It profiles and analyses datasets by single-cell RNA sequencing. Neurodegenerative diseases are identified by the abnormal apoptosis of neurons in the brain with few or no effective therapy strategies at present, which has been a growing healthcare concern and brought a great burden to society. The transcriptome of individual cells provides deep insights into previously unforeseen cellular heterogeneity and gene expression differences in neurodegenerative disorders. It detects multiple cell subsets and functional changes during pathological progression, which deepens the understanding of the molecular underpinnings and cellular basis of neurodegenerative diseases. Furthermore, the transcriptome analysis of immune cells shows the regulation of immune response. Different subtypes of immune cells and their interaction are found to contribute to disease progression. This finding enables the discovery of novel targets and biomarkers for early diagnosis. In this review, we emphasize the principles of the technology, and its recent progress in the study of cellular heterogeneity and immune mechanisms in neurodegenerative diseases. The application of single-cell transcriptomics analysis in neurodegenerative disorders would help explore the pathogenesis of these diseases and develop novel therapeutic methods.
Collapse
Affiliation(s)
- Ziping He
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianqian Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Kaiyue Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiang Lin
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Yilin Peng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Jinlong Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Xisheng Yan
- Department of Cardiovascular Medicine, Wuhan Third Hospital & Tongren Hospital of Wuhan University, Wuhan, China
| | - Yan Jie
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
34
|
Asmis R, Medrano MT, Chase Huizar C, Griffith WP, Forsthuber TG. Dietary Supplementation with 23-Hydroxy Ursolic Acid Reduces the Severity and Incidence of Acute Experimental Autoimmune Encephalomyelitis (EAE) in a Murine Model of Multiple Sclerosis. Nutrients 2024; 16:348. [PMID: 38337633 PMCID: PMC10856865 DOI: 10.3390/nu16030348] [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: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
23-Hydroxy ursolic acid (23-OH UA) is a potent atheroprotective and anti-obesogenic phytochemical, with anti-inflammatory and inflammation-resolving properties. In this study, we examined whether dietary 23-OH UA protects mice against the acute onset and progression of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). Female C57BL/6 mice were fed either a defined low-calorie maintenance diet (MD) or an MD supplemented with 0.2% wgt/wgt 23-OH UA for 5 weeks prior to actively inducing EAE and during the 30 days post-immunization. We observed no difference in the onset of EAE between the groups of mice, but ataxia and EAE disease severity were suppressed by 52% and 48%, respectively, and disease incidence was reduced by over 49% in mice that received 23-OH UA in their diet. Furthermore, disease-associated weight loss was strikingly ameliorated in 23-OH UA-fed mice. ELISPOT analysis showed no significant differences in frequencies of T cells producing IL-17 or IFN-γ between 23-OH UA-fed mice and control mice, suggesting that 23-OH UA does not appear to regulate peripheral T cell responses. In summary, our findings in EAE mice strongly suggest that dietary 23-OH UA may represent an effective oral adjunct therapy for the prevention and treatment of relapsing-remitting MS.
Collapse
Affiliation(s)
- Reto Asmis
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Megan T. Medrano
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA; (M.T.M.)
| | - Carol Chase Huizar
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA; (M.T.M.)
| | - Wendell P. Griffith
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA; (M.T.M.)
| |
Collapse
|
35
|
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
|
36
|
Hu F, Zhu Y, Tian J, Xu H, Xue Q. Single-Cell Sequencing Combined with Transcriptome Sequencing Constructs a Predictive Model of Key Genes in Multiple Sclerosis and Explores Molecular Mechanisms Related to Cellular Communication. J Inflamm Res 2024; 17:191-210. [PMID: 38226354 PMCID: PMC10788626 DOI: 10.2147/jir.s442684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/17/2024] Open
Abstract
Background Multiple sclerosis (MS) causes chronic inflammation and demyelination of the central nervous system and comprises a class of neurodegenerative diseases in which interactions between multiple immune cell types mediate the involvement of MS development. However, the early diagnosis and treatment of MS remain challenging. Methods Gene expression profiles of MS patients were obtained from the Gene Expression Omnibus (GEO) database. Single-cell and intercellular communication analyses were performed to identify candidate gene sets. Predictive models were constructed using LASSO regression. Relationships between genes and immune cells were analyzed by single sample gene set enrichment analysis (ssGSEA). The molecular mechanisms of key genes were explored using gene enrichment analysis. An miRNA network was constructed to search for target miRNAs related to key genes, and related transcription factors were searched by transcriptional regulation analysis. We utilized the GeneCard database to detect the correlations between disease-regulated genes and key genes. We verified the mRNA expression of 4 key genes by reverse transcription-quantitative PCR (RT‒qPCR). Results Monocyte marker genes were selected as candidate gene sets. CD3D, IL2RG, MS4A6A, and NCF2 were found to be the key genes by LASSO regression. We constructed a prediction model with AUC values of 0.7569 and 0.719. The key genes were closely related to immune factors and immune cells. We explored the signaling pathways and molecular mechanisms involving the key genes by gene enrichment analysis. We obtained and visualized the miRNAs associated with the key genes using the miRcode database. We also predicted the transcription factors involved. We used validated key genes in MS patients, several of which were confirmed by RT‒qPCR. Conclusion The prediction model constructed with the CD3D, IL2RG, MS4A6A, and NCF2 genes has good diagnostic efficacy and provides new ideas for the diagnosis and treatment of MS.
Collapse
Affiliation(s)
- Fangzhou Hu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, People’s Republic of China
| | - Yunfei Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, People’s Republic of China
| | - Jingluan Tian
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, People’s Republic of China
| | - Hua Xu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, People’s Republic of China
- Department of Neurology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People’s Hospital, Changzhou, Jiangsu, 215006, People’s Republic of China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, People’s Republic of China
| |
Collapse
|
37
|
Räuber S, Förster M, Schüller J, Willison A, Golombeck KS, Schroeter CB, Oeztuerk M, Jansen R, Huntemann N, Nelke C, Korsen M, Fischer K, Kerkhoff R, Leven Y, Kirschner P, Kölsche T, Nikolov P, Mehsin M, Marae G, Kokott A, Pul D, Schulten J, Vogel N, Ingwersen J, Ruck T, Pawlitzki M, Meuth SG, Melzer N, Kremer D. The Use of Nitrosative Stress Molecules as Potential Diagnostic Biomarkers in Multiple Sclerosis. Int J Mol Sci 2024; 25:787. [PMID: 38255863 PMCID: PMC10815836 DOI: 10.3390/ijms25020787] [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: 12/02/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) of still unclear etiology. In recent years, the search for biomarkers facilitating its diagnosis, prognosis, therapy response, and other parameters has gained increasing attention. In this regard, in a previous meta-analysis comprising 22 studies, we found that MS is associated with higher nitrite/nitrate (NOx) levels in the cerebrospinal fluid (CSF) compared to patients with non-inflammatory other neurological diseases (NIOND). However, many of the included studies did not distinguish between the different clinical subtypes of MS, included pre-treated patients, and inclusion criteria varied. As a follow-up to our meta-analysis, we therefore aimed to analyze the serum and CSF NOx levels in clinically well-defined cohorts of treatment-naïve MS patients compared to patients with somatic symptom disorder. To this end, we analyzed the serum and CSF levels of NOx in 117 patients (71 relapsing-remitting (RR) MS, 16 primary progressive (PP) MS, and 30 somatic symptom disorder). We found that RRMS and PPMS patients had higher serum NOx levels compared to somatic symptom disorder patients. This difference remained significant in the subgroup of MRZ-negative RRMS patients. In conclusion, the measurement of NOx in the serum might indeed be a valuable tool in supporting MS diagnosis.
Collapse
Affiliation(s)
- Saskia Räuber
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Moritz Förster
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology, Kliniken Maria Hilf GmbH, Academic Teaching Hospital of the RWTH Aachen University Hospital, 41063 Moenchengladbach, Germany
| | - Julia Schüller
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Alice Willison
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Kristin S. Golombeck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Christina B. Schroeter
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Menekse Oeztuerk
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Robin Jansen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Melanie Korsen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Katinka Fischer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Ruth Kerkhoff
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology, Kliniken Maria Hilf GmbH, Academic Teaching Hospital of the RWTH Aachen University Hospital, 41063 Moenchengladbach, Germany
| | - Yana Leven
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Patricia Kirschner
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Tristan Kölsche
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Petyo Nikolov
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Mohammed Mehsin
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Gelenar Marae
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Alma Kokott
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Duygu Pul
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Julius Schulten
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Niklas Vogel
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - Nico Melzer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
| | - David Kremer
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (S.R.); (M.F.); (J.S.); (A.W.); (K.S.G.); (C.B.S.); (M.O.); (R.J.); (N.H.); (C.N.); (M.K.); (K.F.); (R.K.); (Y.L.); (P.K.); (T.K.); (P.N.); (G.M.); (A.K.); (D.P.); (J.S.); (N.V.); (J.I.); (T.R.); (M.P.); (S.G.M.); (N.M.)
- Department of Neurology and Neurorehabilitation, Hospital Zum Heiligen Geist, Academic Teaching Hospital of the Heinrich Heine University Düsseldorf, 47906 Kempen, Germany
| |
Collapse
|
38
|
Zhong G, Wang X, Li J, Xie Z, Wu Q, Chen J, Wang Y, Chen Z, Cao X, Li T, Liu J, Wang Q. Insights Into the Role of Copper in Neurodegenerative Diseases and the Therapeutic Potential of Natural Compounds. Curr Neuropharmacol 2024; 22:1650-1671. [PMID: 38037913 PMCID: PMC11284712 DOI: 10.2174/1570159x22666231103085859] [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: 02/07/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 12/02/2023] Open
Abstract
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
39
|
Li N, Han X, Ruan M, Huang F, Yang L, Xu T, Wang H, Wu H, Shi S, Wang Y, Wu X, Wang S. Prebiotic inulin controls Th17 cells mediated central nervous system autoimmunity through modulating the gut microbiota and short chain fatty acids. Gut Microbes 2024; 16:2402547. [PMID: 39287045 PMCID: PMC11409507 DOI: 10.1080/19490976.2024.2402547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination occurring in the central nervous system (CNS). Inulin is a common prebiotic that can improve metabolic disorders by modulating the gut microbiota. However, its capacity to affect CNS autoimmunity is poorly recognized. Experimental autoimmune encephalomyelitis (EAE) is a classical mouse model of MS. Herein, we found that oral administration of inulin ameliorated the severity EAE in mice, accompanied by reductions in inflammatory cell infiltration and demyelination in the CNS. These reductions were associated with decreased proportion and numbers of Th17 cells in brain and spleen. Consistent with the findings, the serum concentrations of IL-17, IL-6, and TNF-α were reduced in inulin treated EAE mice. Moreover, the proliferation of auto-reactive lymphocytes, against MOG35-55 antigen, was attenuated ex vivo. Mechanistically, inulin treatment altered the composition of gut microbiota. It increased Lactobacillus and Dubosiella whereas decreased g_Prevotellaceae_NK3B31_group at the genus level, alongside with elevated concentration of butyric acid in fecal content and serum. In vitro, butyrate, but not inulin, could inhibit the activation of MOG35-55 stimulated lymphocytes. Furthermore, fecal microbiota transplantation assay confirmed that fecal contents of inulin-treated normal mice had an ameliorative effect on EAE mice. In contrast, antibiotic cocktail (ABX) treatment diminished the therapeutic effect of inulin in EAE mice as well as the reduction of Th17 cells, while supplementation with Lactobacillus reuteri restored the amelioration effect. These results confirmed that the attenuation of inulin on Th17 cells and inflammatory demyelination in EAE mice was dependent on its modulation on gut microbiota and metabolites. Our findings provide a potential therapeutic regimen for prebiotic inulin supplementation in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Ning Li
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyan Han
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Songshan Shi
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Mendes O. Inflammation and neurodegeneration in multiple sclerosis. A REVIEW ON DIVERSE NEUROLOGICAL DISORDERS 2024:321-345. [DOI: 10.1016/b978-0-323-95735-9.00023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Neziraj T, Siewert L, Pössnecker E, Pröbstel AK. Therapeutic targeting of gut-originating regulatory B cells in neuroinflammatory diseases. Eur J Immunol 2023; 53:e2250033. [PMID: 37624875 DOI: 10.1002/eji.202250033] [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: 02/06/2023] [Revised: 05/29/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
Regulatory B cells (Bregs) are immunosuppressive cells that support immunological tolerance by the production of IL-10, IL-35, and TGF-β. Bregs arise from different developmental stages in response to inflammatory stimuli. In that regard, mounting evidence points towards a direct influence of gut microbiota on mucosal B cell development, activation, and regulation in health and disease. While an increasing number of diseases are associated with alterations in gut microbiome (dysbiosis), little is known about the role of microbiota on Breg development and induction in neuroinflammatory disorders. Notably, gut-originating, IL-10- and IgA-producing regulatory plasma cells have recently been demonstrated to egress from the gut to suppress inflammation in the CNS raising fundamental questions about the triggers and functions of mucosal-originating Bregs in systemic inflammation. Advancing our understanding of Bregs in neuroinflammatory diseases could lead to novel therapeutic approaches. Here, we summarize the main aspects of Breg differentiation and functions and evidence about their involvement in neuroinflammatory diseases. Further, we highlight current data of gut-originating Bregs and their microbial interactions and discuss future microbiota-regulatory B cell-targeted therapies in immune-mediated diseases.
Collapse
Affiliation(s)
- Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Lena Siewert
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
42
|
Tortosa-Carreres J, Quiroga-Varela A, Castillo-Villalba J, Piqueras-Rodríguez M, Ramió-Torrenta L, Cubas-Núñez L, Gasqué-Rubio R, Quintanilla-Bordas C, Huertas-Pons JM, Miguela A, Casanova B, Laiz-Marro B, Pérez-Miralles FC. Improving the efficiency of free kappa light chains as diagnostic biomarker of Multiple Sclerosis by using a novel algorithm. Mult Scler Relat Disord 2023; 79:104997. [PMID: 37714099 DOI: 10.1016/j.msard.2023.104997] [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: 08/01/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Intrathecal immunoglobulin synthesis (ITS) plays a crucial role in the diagnosis of multiple sclerosis (MS). Traditionally, the gold standard method for detecting ITS has been through the analysis of oligoclonal bands (OCB). However, the paradigm has shifted with the introduction of the free kappa light chains (FKLC) method. In fact, a recent consensus recommends evaluating FKLC index (FKLCi) as the primary approach and reserving oligoclonal bands with borderline results. The objective of our study is to investigate the diagnostic efficiency of combining FKLC with other methods to predict ITS while minimizing the reliance on OCB. METHODS A total of 192 patients were included in the study, consisting of 145 individuals diagnosed with multiple sclerosis (pwMS) and 46 with other neurological diseases (controls). Among the MS cases, 100 patients were assigned to the Training Cohort (TC), while an external Validation Cohort (VC) comprised of 45 MS patients was established. Diagnostic efficiency was assessed for FKLCi, OCB, Link index, and the Reiber formula for IgG and FKLC. Optimal cutoff values for Link index and FKLCi were also determined. The last procedure was developed for diverse algorithms using the parameters mentioned above, which included the optimal cutoffs previously obtained. The calculations were conducted independently for both the TC and the VC, as well as for a composite cohort formed by combining data from all patients (OC) RESULTS: One algorithm, named KRO, was developed based on the determination of FKLCi and Reiber Formula as the primary diagnostic parameters. For cases where the FKLCi result was mildly increased, OCB was utilized as a supplementary test. The KRO algorithm demonstrated superior diagnostic accuracy in the OC (89%), resulting in a reduction of OCB consumption by 91%. DISCUSSION The KRO algorithm demonstrated superior sensitivity and accuracy although lower specificity and NPV compared to the use of FKLCi and OCB alone. The present research aligns with the new consensus recommendations regarding the diagnostic approach. Our findings indicate that employing a combined marker approach via KRO could prove to be a proficient screening tool for multiple sclerosis. This approach also holds the potential to address inherent limitations associated with each individual marker. However, to further validate and solidify the efficacy of our algorithm, additional studies involving larger cohorts are warranted.
Collapse
Affiliation(s)
- Jordi Tortosa-Carreres
- Laboratory Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain; Medicine Department, University of Valencia, Valencia 46010, Spain.
| | - Anna Quiroga-Varela
- Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Jessica Castillo-Villalba
- Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España; Medicine Department, University of Valencia, Valencia 46010, Spain
| | - Mónica Piqueras-Rodríguez
- Laboratory Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain; Medicine Department, University of Valencia, Valencia 46010, Spain
| | - Lluís Ramió-Torrenta
- Girona Neuroimmumology and Multiple Sclerosis Unit, Neurology Department, Dr. Josep Trueta University Hospital and Santa Caterina Hospital, Girona, Spain; Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Medical Sciences Department, University of Girona, Girona, Spain
| | - Laura Cubas-Núñez
- Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España
| | - Raquel Gasqué-Rubio
- Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España; Medicine Department, University of Valencia, Valencia 46010, Spain
| | - Carlos Quintanilla-Bordas
- Neurology Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain; Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España
| | - Joana María Huertas-Pons
- Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Albert Miguela
- Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Bonaventura Casanova
- Neurology Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain; Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España
| | - Begoña Laiz-Marro
- Laboratory Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain
| | - Francisco Carlos Pérez-Miralles
- Neurology Department, La Fe University and Polytechnic Hospital, Valencia 46026, Spain; Grupo de investigación en Neuroinmunología, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, España
| |
Collapse
|
43
|
Ogami S, Koh J, Miyamoto K, Mori M, Takahashi M, Nakayama Y, Sakata M, Hiwatani Y, Kajimoto Y, Ishiguchi H, Ito H. Predictive value of the neutrophil-to-lymphocyte ratio for treatment response in patients diagnosed with definite or probable autoimmune encephalitis/encephalopathy. Front Neurol 2023; 14:1284717. [PMID: 37936916 PMCID: PMC10626493 DOI: 10.3389/fneur.2023.1284717] [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: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Autoimmune encephalitis/encephalopathy (AE) is a complex and heterogeneous disease, making it difficult to predict the prognosis. The neutrophil-to-lymphocyte ratio (NLR) has emerged as a potential prognostic tool, but its usefulness remains a matter of debate. This study aimed to explore prognostic factors in cases of clinically definite or probable AE, including those with autoantibody-negative, or unknown status. Methods Data on patients diagnosed with definite or probable AE, including those with autoantibody-negative, or unknown status, were retrospectively collected from the admission records of our department between January 2013 and December 2022. These patients were then categorized into either a good- or poor-response group, based on their short-term treatment response. Clinical characteristics, auxiliary examinations, and treatments were compared between the two groups. A multivariable logistic regression model was constructed to identify independent predictors of poor short-term treatment response by Akaike information criterion backward stepwise method. Results A total of 31 patients were included in the final analysis, with 18 of them included in the poor-response group. In the univariable analysis, the poor-response group had a higher proportion of patients with a modified Rankin Scale (mRS) high score upon admission, female, epileptic seizures, or NLRs of 3.93 or higher than the good-response group (all p < 0.10). Furthermore, the multivariable logistic regression analysis revealed that the mRS score upon admission [OR: 5.51, 95% confidence intervals (CI): 1.29-23.50, p = 0.02], epileptic seizures (OR: 10.01, 95% CI: 1.16-86.66, p = 0.04), and NLRs of 3.93 or higher (OR: 11.37, 95% CI: 1.12-114.68, p = 0.04) were significantly associated with poor short-term treatment response. Conclusion The NLR may play a supplementary role in predicting the short-term treatment response in patients diagnosed with definite or probable AE, including those with autoantibody-negative, or unknown status.
Collapse
Affiliation(s)
- Shuhei Ogami
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Jinsoo Koh
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | | | - Megumi Mori
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Maiko Takahashi
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Yoshiaki Nakayama
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Mayumi Sakata
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Yasuhiro Hiwatani
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
- Department of Neurology, Wakayama Rosai Hospital, Wakayama, Japan
| | | | - Hiroshi Ishiguchi
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
- Department of Neurology, Shingu Municipal Medical Center, Wakayama, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
44
|
Rogić Vidaković M, Ćurković Katić A, Pavelin S, Bralić A, Mikac U, Šoda J, Jerković A, Mastelić A, Dolić K, Markotić A, Đogaš Z, Režić Mužinić N. Transcranial Magnetic Stimulation Measures, Pyramidal Score on Expanded Disability Status Scale and Magnetic Resonance Imaging of Corticospinal Tract in Multiple Sclerosis. Bioengineering (Basel) 2023; 10:1118. [PMID: 37892848 PMCID: PMC10604490 DOI: 10.3390/bioengineering10101118] [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: 09/04/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Probing the cortic ospinal tract integrity by transcranial magnetic stimulation (TMS) could help to understand the neurophysiological correlations of multiple sclerosis (MS) symptoms. Therefore, the study objective was, first, to investigate TMS measures (resting motor threshold-RMT, motor evoked potential (MEP) latency, and amplitude) of corticospinal tract integrity in people with relapsing-remitting MS (pwMS). Then, the study examined the conformity of TMS measures with clinical disease-related (Expanded Disability Status Scale-EDSS) and magnetic resonance imaging (MRI) results (lesion count) in pwMS. The e-field navigated TMS, MRI, and EDSS data were collected in 23 pwMS and compared to non-clinical samples. The results show that pwMS differed from non-clinical samples in MEP latency for upper and lower extremity muscles. Also, pwMS with altered MEP latency (prolonged or absent MEP response) had higher EDSS, general and pyramidal, functional scores than pwMS with normal MEP latency finding. Furthermore, the RMT intensity for lower extremity muscles was predictive of EDSS functional pyramidal scores. TMS/MEP latency findings classified pwMS as the same as EDSS functional pyramidal scores in 70-83% of cases and were similar to the MRI results, corresponding to EDSS functional pyramidal scores in 57-65% of cases. PwMS with altered MEP latency differed from pwMS with normal MEP latency in the total number of lesions in the brain corticospinal and cervical corticospinal tract. The study provides preliminary results on the correspondence of MRI and TMS corticospinal tract evaluation results with EDSS functional pyramidal score results in MS.
Collapse
Affiliation(s)
- Maja Rogić Vidaković
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia; (A.J.); (Z.Đ.)
| | - Ana Ćurković Katić
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia;
| | - Sanda Pavelin
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia;
| | - Antonia Bralić
- Department of Interventional and Diagnostic Radiology, University Hospital of Split, 21000 Split, Croatia; (A.B.); (K.D.)
| | - Una Mikac
- Department of Psychology, Faculty of Humanities and Social Sciences University of Zagreb, 10000 Zagreb, Croatia;
| | - Joško Šoda
- Signal Processing, Analysis, Advanced Diagnostics Research and Education Laboratory (SPAADREL), Faculty of Maritime Studies, Department for Marine Electrical Engineering and Information Technologies, University of Split, 21000 Split, Croatia;
| | - Ana Jerković
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia; (A.J.); (Z.Đ.)
| | - Angela Mastelić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.); (A.M.); (N.R.M.)
| | - Krešimir Dolić
- Department of Interventional and Diagnostic Radiology, University Hospital of Split, 21000 Split, Croatia; (A.B.); (K.D.)
- Department of Radiology, School of Medicine, University of Split, 21000 Split, Croatia
| | - Anita Markotić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.); (A.M.); (N.R.M.)
| | - Zoran Đogaš
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia; (A.J.); (Z.Đ.)
| | - Nikolina Režić Mužinić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia; (A.M.); (A.M.); (N.R.M.)
| |
Collapse
|
45
|
Long Z, Zeng L, He Q, Yang K, Xiang W, Ren X, Deng Y, Chen H. Research progress on the clinical application and mechanism of iguratimod in the treatment of autoimmune diseases and rheumatic diseases. Front Immunol 2023; 14:1150661. [PMID: 37809072 PMCID: PMC10552782 DOI: 10.3389/fimmu.2023.1150661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/04/2023] [Indexed: 10/10/2023] Open
Abstract
Autoimmune diseases are affected by complex pathophysiology involving multiple cell types, cytokines, antibodies and mimicking factors. Different drugs are used to improve these autoimmune responses, including nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, antibodies, and small molecule drugs (DMARDs), which are prevalent clinically in the treatment of rheumatoid arthritis (RA), etc. However, low cost-effectiveness, reduced efficacy, adverse effects, and patient non-response are unattractive factors driving the development of new drugs such as iguratimod. As a new disease-modifying antirheumatic drug, iguratimod has pharmacological activities such as regulating autoimmune disorders, inflammatory cytokines, regulating immune cell activation, differentiation and proliferation, improving bone metabolism, and inhibiting fibrosis. In recent years, clinical studies have found that iguratimod is effective in the treatment of RA, SLE, IGG4-RD, Sjogren 's syndrome, ankylosing spondylitis, interstitial lung disease, and other autoimmune diseases and rheumatic diseases. The amount of basic and clinical research on other autoimmune diseases is also increasing. Therefore, this review systematically reviews the latest relevant literature in recent years, reviews the research results in recent years, and summarizes the research progress of iguratimod in the treatment of related diseases. This review highlights the role of iguratimod in the protection of autoimmune and rheumatic bone and related immune diseases. It is believed that iguratimod's unique mode of action and its favorable patient response compared to other DMARDs make it a suitable antirheumatic and bone protective agent in the future.
Collapse
Affiliation(s)
- Zhiyong Long
- Department of Rehabilitation Medicine, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, Hunan, China
| | - Xiang Ren
- Department of Rheumatology, The First People's Hospital Changde City, Changde, Hunan, China
| | - Ying Deng
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Hua Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
46
|
Brune-Ingebretsen S, Høgestøl EA, de Rosbo NK, Berg-Hansen P, Brunborg C, Blennow K, Zetterberg H, Paul F, Uccelli A, Villoslada P, Harbo HF, Berge T. Immune cell subpopulations and serum neurofilament light chain are associated with increased risk of disease worsening in multiple sclerosis. J Neuroimmunol 2023; 382:578175. [PMID: 37573634 DOI: 10.1016/j.jneuroim.2023.578175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/18/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Changes is lymphocyte subpopulations in peripheral blood have been proposed as biomarkers for evaluation of disease activity in multiple sclerosis (MS). Serum neurofilament light chain (sNfL) is a biomarker reflecting neuro-axonal injury in MS that could be used to monitor disease activity, response to drugs and to prognosticate disease course. Here we show a moderate correlation between sNfL and lymphocyte cell subpopulations, and our data furthermore suggest that sNfL and specific immune cell subpopulations together could predict future disease worsening in MS.
Collapse
Affiliation(s)
- Synne Brune-Ingebretsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway.
| | - Einar A Høgestøl
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Nicole Kerlero de Rosbo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; TomaLab, Institute of Nanotechnology, National Research Council (CNR), Rome, Italy
| | - Pål Berg-Hansen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitaetsmedizin Berlin, Berlin, Germany; NeuroCure Clinical Research Center, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Pablo Villoslada
- Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Hanne F Harbo
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Tone Berge
- Department of Research, Innovation and Education, Oslo University Hospital, Oslo, Norway; Department of Mechanical, Electronic and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
47
|
Xiong R, Lei J, Pan S, Zhang H, Tong Y, Wu W, Huang Y, Lai X. Post-marketing safety surveillance of dalfampridine for multiple sclerosis using FDA adverse event reporting system. Front Pharmacol 2023; 14:1226086. [PMID: 37781705 PMCID: PMC10538962 DOI: 10.3389/fphar.2023.1226086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Objective: To investigate and analyze the post-marketing adverse event (AE) data of multiple sclerosis (MS) therapeutic drug dalfampridine using the US Food and Drug Administration Adverse Event Reporting System (FAERS) for its clinical safety application. Methods: Use OpenVigil2.1 platform to obtain AE data of dalfampridine from FAERS from February 2010 to September 2022. Match "adverse drug reaction" with preferred term (PT) and system organ class (SOC) according to the Medical Dictionary for Regulatory Activities (MedDRA), then merge the same PT and delete non-AE PT. Positive signals were identified by the reporting odds ratio (ROR), proportional reporting ratio (PRR), and Bayesian confidence propagation neural network (BCPNN) methods. When AE signals met the criteria of those three methods, they were identified as positive signals. Results: A total of 44,092 dalfampridine-related AE reports were obtained, and 335 AE signals were identified, including 11,889 AE reports. AEs were more common in females and in the 45-65 age group, which is consistent with the epidemiological characteristics of MS. The 335 AE signals involved 21 SOCs, including investigations, infections and infestations, eye disorders, etc. Among the top 20 PTs in signal strength, 10 were associated with abnormal lymphocyte percentage and count, and 5 were associated with abnormal urine tests, some of which were not described in the instruction, such as spinal cord injury cauda equina, haemoglobin urine present, urinary sediment abnormal and so on. The most frequently reported AE signals were urinary tract infection, dizziness, condition aggravated. In addition, 23 AE signals with death outcomes were identified, with an incidence of less than 0.1%. Conclusion: Data mining of FAERS was conducted to analyze the AEs of dalfampridine, and new AE signals were found. This study provides a reference for the safe use of dalfampridine in the treatment of MS.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Pharmacy, The 958th Hospital of Chinese People’s Liberation Army, Chongqing, China
| | - Jing Lei
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Sicen Pan
- Department of Pharmacy, The 958th Hospital of Chinese People’s Liberation Army, Chongqing, China
| | - Hong Zhang
- Department of Pharmacy, The 956th Hospital of Chinese People’s Liberation Army, Nyingchi, Tibet, China
| | - Yongtao Tong
- Department of Pharmacy, The 956th Hospital of Chinese People’s Liberation Army, Nyingchi, Tibet, China
| | - Wei Wu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Xiaodan Lai
- Department of Pharmacy, The 958th Hospital of Chinese People’s Liberation Army, Chongqing, China
| |
Collapse
|
48
|
Zhang S, Ma Y, Luo X, Xiao H, Cheng R, Jiang A, Qin X. Integrated Analysis of Immune Infiltration and Hub Pyroptosis-Related Genes for Multiple Sclerosis. J Inflamm Res 2023; 16:4043-4059. [PMID: 37727371 PMCID: PMC10505586 DOI: 10.2147/jir.s422189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/02/2023] [Indexed: 09/21/2023] Open
Abstract
Purpose Studies on overall immune infiltration and pyroptosis in patients with multiple sclerosis (MS) are limited. This study explored immune cell infiltration and pyroptosis in MS using bioinformatics and experimental validation. Methods The GSE131282 and GSE135511 microarray datasets including brain autopsy tissues from controls and MS patients were downloaded for bioinformatic analysis. The gene expression-based deconvolution method, CIBERSORT, was used to determine immune infiltration. Differentially expressed genes (DEGs) and functional enrichments were analyzed. We then extracted pyroptosis-related genes (PRGs) from the DEGs by using machine learning strategies. Their diagnostic ability for MS was evaluated in both the training set (GSE131282 dataset) and validation set (GSE135511 dataset). In addition, messenger RNA (mRNA) expression of PRGs was validated using quantitative real-time polymerase chain reaction (qRT-PCR) in cortical tissue from an experimental autoimmune encephalomyelitis (EAE) model of MS. Moreover, the functional enrichment pathways of each hub PRG were estimated. Finally, co-expressed competitive endogenous RNA (ceRNA) networks of PRGs in MS were constructed. Results Among the infiltrating cells, naive CD4+ T cells (P=0.006), resting NK cells (P=0.002), activated mast cells (P=0.022), and neutrophils (P=0.002) were significantly higher in patients with MS than in controls. The DEGs of MS were screened. Analysis of enrichment pathways showed that the pathways of transcriptional regulatory mechanisms and ion channels associating with pyroptosis. Four PRGs genes CASP4, PLCG1, CASP9 and NLRC4 were identified. They were validated in both the GSE135511 dataset and the EAE model by using qRT-PCR. CASP4 and NLRC4 were ultimately identified as stable hub PRGs for MS. Single-gene Gene Set Enrichment Analysis showed that they mainly participated in biosynthesis, metabolism, and organism resistance. ceRNA networks containing CASP4 and NLRC4 were constructed. Conclusion MS was associated with immune infiltration. CASP4 and NLRC4 were key biomarkers of pyroptosis in MS.
Collapse
Affiliation(s)
- Shaoru Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yue Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaoqin Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongmei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ruiqi Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Anan Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
49
|
Louapre C, Rosenzwajg M, Golse M, Roux A, Pitoiset F, Adda L, Tchitchek N, Papeix C, Maillart E, Ungureanu A, Charbonnier-Beaupel F, Galanaud D, Corvol JC, Vicaut E, Lubetzki C, Klatzmann D. A randomized double-blind placebo-controlled trial of low-dose interleukin-2 in relapsing-remitting multiple sclerosis. J Neurol 2023; 270:4403-4414. [PMID: 37245191 DOI: 10.1007/s00415-023-11690-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is associated with regulatory T cells (Tregs) insufficiency while low-dose interleukin-2 (IL2LD) activates Tregs and reduces disease activity in autoimmune diseases. METHODS We aimed at addressing whether IL2LD improved Tregs from MS patients. MS-IL2 was a single-center double-blind phase-2 study. Thirty patients (mean [SD] age 36.8 years [8.3], 16 female) with relapsing-remitting MS with new MRI lesions within 6 months before inclusion were randomly assigned in a 1:1 ratio to placebo or IL-2 at 1 million IU, daily for 5 days and then fortnightly for 6 months. The primary endpoint was change in Tregs at day-5. RESULTS Unlike previous trials of IL2LD in more than 20 different autoimmune diseases, Tregs were not expanded at day-5 in IL2LD group, but only at day-15 (median [IQR] fold change from baseline: 1.26 [1.21-1.33] in IL2LD group; 1.01 [0.95-1.05] in placebo group, p < 0.001). At day-5, however, Tregs had acquired an activated phenotype (fold change of CD25 expression in Tregs: 2.17 [1.70-3.55] in IL2LD versus 0.97 [0.86-1.28] in placebo group, p < 0.0001). Regulator/effector T cells ratio remained elevated throughout treatment period in the IL2LD group (p < 0.001). Number of new active brain lesions and of relapses tended to be reduced in IL2LD treated patients, but the difference did not reach significance in this trial not powered to detect clinical efficacy. CONCLUSION The effect of IL2LD on Tregs in MS patients was modest and delayed, compared to other auto-immune diseases. This, together with findings that Tregs improve remyelination in MS models and recent reports of IL2LD efficacy in amyotrophic lateral sclerosis, warrants larger studies of IL2LD in MS, notably with increased dosages and/or modified modalities of administration. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov: NCT02424396; EU Clinical trials Register: 2014-000088-42.
Collapse
Affiliation(s)
- C Louapre
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - M Rosenzwajg
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
| | - M Golse
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - A Roux
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
| | - F Pitoiset
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
| | - L Adda
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France
| | - N Tchitchek
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France
| | - C Papeix
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - E Maillart
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - A Ungureanu
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - F Charbonnier-Beaupel
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Pharmacie à Usage Intérieur, Reqpharm Unit, Paris, France
| | - D Galanaud
- Neuroradiology Department, Sorbonne University, Assistance Publique Hôpitaux de Paris, Paris, France
| | - J C Corvol
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - E Vicaut
- Assistance Publique Hôpitaux de Paris, Lariboisière Hospital, Clinical Trial Unit, Paris, France
| | - C Lubetzki
- Sorbonne University, Paris Brain Institute - ICM, Assistance Publique Hôpitaux de Paris, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Department of Neurology, CIC neurosciences, Paris, France
| | - D Klatzmann
- Immunology-Immunopathology-Immunotherapy (i3)-UMRS_959, Sorbonne Université- INSERM, Paris, France.
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Clinical Investigation Center for Biotherapies (CIC-BTi) and Immunology-Inflammation-Infectiology and Dermatology Department (3iD), Paris, France.
| |
Collapse
|
50
|
Xavier A, Maltby VE, Ewing E, Campagna MP, Burnard SM, Tegner JN, Slee M, Butzkueven H, Kockum I, Kular L, Jokubaitis VG, Kilpatrick T, Alfredsson L, Jagodic M, Ponsonby AL, Taylor BV, Scott RJ, Lea RA, Lechner-Scott J. DNA Methylation Signatures of Multiple Sclerosis Occur Independently of Known Genetic Risk and Are Primarily Attributed to B Cells and Monocytes. Int J Mol Sci 2023; 24:12576. [PMID: 37628757 PMCID: PMC10454485 DOI: 10.3390/ijms241612576] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Epigenetic mechanisms can regulate how DNA is expressed independently of sequence and are known to be associated with various diseases. Among those epigenetic mechanisms, DNA methylation (DNAm) is influenced by genotype and the environment, making it an important molecular interface for studying disease etiology and progression. In this study, we examined the whole blood DNA methylation profiles of a large group of people with (pw) multiple sclerosis (MS) compared to those of controls. We reveal that methylation differences in pwMS occur independently of known genetic risk loci and show that they more strongly differentiate disease (AUC = 0.85, 95% CI 0.82-0.89, p = 1.22 × 10-29) than known genetic risk loci (AUC = 0.72, 95% CI: 0.66-0.76, p = 9.07 × 10-17). We also show that methylation differences in MS occur predominantly in B cells and monocytes and indicate the involvement of cell-specific biological pathways. Overall, this study comprehensively characterizes the immune cell-specific epigenetic architecture of MS.
Collapse
Affiliation(s)
- Alexandre Xavier
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Vicki E. Maltby
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Sean M. Burnard
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Jesper N. Tegner
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia;
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, 17176 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, 17165 Solna, Sweden
| | - Mark Slee
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
- MSBase Foundation, Melbourne, VIC 3004, Australia
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | | | - Vilija G. Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Trevor Kilpatrick
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
| | - Lars Alfredsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
- National Centre for Epidemiology and Public Health, Australian National University, Canberra, ACT 2601, Australia
| | - Bruce V. Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia;
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
- Department of Molecular Genetics, Pathology North, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Rodney A. Lea
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Jeannette Lechner-Scott
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|