1
|
Wiendl H, Barkhof F, Montalban X, Achiron A, Derfuss T, Chan A, Hodgkinson S, Prat A, Leocani L, Schmierer K, Sellebjerg F, Vermersch P, Jin H, Chudecka A, Kloetgen A, Lin D, Gardner L, De Stefano N. Blood biomarker dynamics in people with relapsing multiple sclerosis treated with cladribine tablets: results of the 2-year MAGNIFY-MS study. Front Immunol 2025; 16:1512189. [PMID: 39963134 PMCID: PMC11830603 DOI: 10.3389/fimmu.2025.1512189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
Background and objectives Cladribine tablets (CladT) represent an effective immune reconstitution therapy, administered in short treatment courses over two consecutive years. To better understand the amplitude of immune changes, we performed a comprehensive analysis during the 2-year study period for the entire MAGNIFY-MS population (N=270). In addition to lymphocyte kinetics, we studied intracellular cytokines serum proteins, and their associations with clinical outcomes. To put these changes into perspective, we analyzed transcriptional changes in T and B cells and associated biological pathways before and after each treatment course with CladT. Methods Immunophenotyping and transcriptomics were performed at regular visits with major differences reported between baseline (BL) and after each yearly treatment course. Assessments included: lymphocyte dynamics, RNA sequencing (B and T cells), intracellular cytokines, serum proteins (immunoglobulins [IgG and IgM], and serum neurofilament light chain [sNfL]). Clinical measures included: MRI activity, annualized relapse rate (ARR), 6-month confirmed disability progression (6mCDP), timed 25-foot walk (T25FW), and 9-hole peg test (9HPT). Results All B, T and NK cells were reduced at month (M)3 after CladT administration, except regulatory B cells which increased above BL from M3 to M24. Naïve and transitional B cells recovered at M6; all other B and T cell subsets remained below BL levels. Reductions in all NK cell subtypes were observed at M3, CD16lowCD56bright and NKp46 cells reconstituted at M6 and M12 respectively. Changes in genes and pathways associated with innate and adaptive immune response were observed after CladT treatment, along with reductions in pro-inflammatory cytokine-producing B and T cells and increases in anti-inflammatory cytokine-producing T cells. IgG and IgM levels remained above the lower limits of normal in most participants. sNfL levels decreased, remaining reduced by M24. Significant reductions in the annualized combined unique active lesion count occurred from M2 onwards. ARR was 0.11 (95% confidence interval: 0.09,0.15), with 83% participants free of qualifying relapses. Over 90% of participants were free of 6mCDP, around 87% had no confirmed progression on T25FW and 9HPT. No significant correlations were seen between clinical parameters and lymphocyte dynamics to M6. The safety profile was consistent with previous reports. Discussion Deep longitudinal immunophenotyping, analysis of transcriptional changes, reduction in cells expressing pro-inflammatory cytokines, along with the marker of neuroaxonal damage provide novel and innovative evidence of CladT rebalancing the immune system towards a more homeostatic and less pathogenic state. Clinical Trial Registration https://clinicaltrials.gov/study/, identifier NCT03364036.
Collapse
Affiliation(s)
- Heinz Wiendl
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Xavier Montalban
- Department of Neurology-Neuroimmunology, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Hospital Universitario Vall d’Hebron, Barcelona, Spain
| | - Anat Achiron
- Multiple Sclerosis Center, Sheba Academic Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tobias Derfuss
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Suzanne Hodgkinson
- Ingham Institute for Applied Medical Research, University of New South Wales Medicine and Liverpool Hospital, Sydney, NSW, Australia
| | - Alexandre Prat
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Letizia Leocani
- Department of Neurology, University Vita-Salute San Raffaele, Milan, Italy
- Experimental Neurophysiology Unit, Scientific Institute IRCCS San Raffaele, Milan, Italy
- Department of Neurorehabilitation Science, Casa di Cura Igea, Milan, Italy
| | - Klaus Schmierer
- The Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS, Trust, London, United Kingdom
| | - Finn Sellebjerg
- Danish MS Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Patrick Vermersch
- Univ. Lille, Inserm U1172 LilNCog, CHU Lille, FHU Precise, Lille, France
| | - Hulin Jin
- Clinical Measurement Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Anita Chudecka
- Clinical Research Services, Cytel Inc., Geneva, Switzerland
| | - Andreas Kloetgen
- Clinical Measurement Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Dongdong Lin
- Clinical Measurement Sciences, EMD Serono Research & Development Institute, Inc., an affiliate of Merck KGaA, Billerica, MA, United States
| | - Lidia Gardner
- Neurology & Immunology Medical Unit, EMD Serono Research & Development Institute, Inc., an affiliate of Merck KGaA, Billerica, MA, United States
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
2
|
de Weerd HA, Guala D, Gustafsson M, Synnergren J, Tegnér J, Lubovac-Pilav Z, Magnusson R. Latent space arithmetic on data embeddings from healthy multi-tissue human RNA-seq decodes disease modules. PATTERNS (NEW YORK, N.Y.) 2024; 5:101093. [PMID: 39568475 PMCID: PMC11573900 DOI: 10.1016/j.patter.2024.101093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Computational analyses of transcriptomic data have dramatically improved our understanding of complex diseases. However, such approaches are limited by small sample sets of disease-affected material. We asked if a variational autoencoder trained on large groups of healthy human RNA sequencing (RNA-seq) data can capture the fundamental gene regulation system and generalize to unseen disease changes. Importantly, we found this model to successfully compress unseen transcriptomic changes from 25 independent disease datasets. We decoded disease-specific signals from the latent space and found them to contain more disease-specific genes than the corresponding differential expression analysis in 20 of 25 cases. Finally, we matched these disease signals with known drug targets and extracted sets of known and potential pharmaceutical candidates. In summary, our study demonstrates how data-driven representation learning enables the arithmetic deconstruction of the latent space, facilitating the dissection of disease mechanisms and drug targets.
Collapse
Affiliation(s)
- Hendrik A de Weerd
- School of Bioscience, Systems Biology Research Center, University of Skövde, 541 45 Skövde, Sweden
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
- Department of Biomedical Engineering, Linköping University, 581 83 Linköping, Sweden
| | - Dimitri Guala
- Department of Biochemistry and Biophysics, Stockholm University, 171 21 Solna, Sweden
- Merck AB, 169 70 Solna, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Jane Synnergren
- School of Bioscience, Systems Biology Research Center, University of Skövde, 541 45 Skövde, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Jesper Tegnér
- Biological and Environmental Science 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, 171 76, Stockholm, Sweden
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Solna, Sweden
| | - Zelmina Lubovac-Pilav
- School of Bioscience, Systems Biology Research Center, University of Skövde, 541 45 Skövde, Sweden
| | - Rasmus Magnusson
- School of Bioscience, Systems Biology Research Center, University of Skövde, 541 45 Skövde, Sweden
- Department of Biomedical Engineering, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
3
|
Rodrigues Barreto I, Monteiro A, Paiva A, Fonseca AM. Relapsing-remitting multiple sclerosis patients exhibit differential natural killer functional subpopulations. Acta Neurol Belg 2024; 124:603-610. [PMID: 38441808 DOI: 10.1007/s13760-024-02488-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/22/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) and has been known as T-cell mediated. However, the contribution of multiple cell types, notably natural killer (NK) cells, has also been reported. AIM To quantify circulating total NK cells and its subpopulations, CD56 dim and bright, and to characterize the functional phenotype and IFN-γ and TNF-α production in relapsing-remitting patients treated with IFN-β and in apparently healthy controls. RESULTS CD56bright NK cells were found to be the least represented subpopulation. In relapse patients, the frequencies of IFN-γ-producing NK cells and their subpopulations were significantly decreased. In remission patients, CD56dim NK cells expressed high levels of HLA-DR and CD54. CONCLUSION These results suggest that remission RRMS patients, although in an inactive stage of MS, present circulating NK cells with an activation phenotype, supporting the idea that NK cells may be relevant mediators in the MS pathophysiology.
Collapse
Affiliation(s)
- Inês Rodrigues Barreto
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Andreia Monteiro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
- Clinical Pathology Service, Centro Hospitalar Universitário da Cova da Beira (CHUCB), Covilhã, Portugal
| | - Artur Paiva
- Cytometry Operational Management Unit, Clinical Pathology Service, CHUC, Coimbra, Portugal
- iCBR - Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal
- ESTESC-Coimbra Health School, Coimbra, Portugal
| | - Ana Mafalda Fonseca
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
| |
Collapse
|
4
|
Grunwald C, Krętowska-Grunwald A, Adamska-Patruno E, Kochanowicz J, Kułakowska A, Chorąży M. The Role of Selected Interleukins in the Development and Progression of Multiple Sclerosis-A Systematic Review. Int J Mol Sci 2024; 25:2589. [PMID: 38473835 PMCID: PMC10932438 DOI: 10.3390/ijms25052589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Multiple sclerosis is a disabling inflammatory disorder of the central nervous system characterized by demyelination and neurodegeneration. Given that multiple sclerosis remains an incurable disease, the management of MS predominantly focuses on reducing relapses and decelerating the progression of both physical and cognitive decline. The continuous autoimmune process modulated by cytokines seems to be a vital contributing factor to the development and relapse of multiple sclerosis. This review sought to summarize the role of selected interleukins in the pathogenesis and advancement of MS. Patients with MS in the active disease phase seem to exhibit an increased serum level of IL-2, IL-4, IL-6, IL-13, IL-17, IL-21, IL-22 and IL-33 compared to healthy controls and patients in remission, while IL-10 appears to have a beneficial impact in preventing the progression of the disease. Despite being usually associated with proinflammatory activity, several studies have additionally recognized a neuroprotective role of IL-13, IL-22 and IL-33. Moreover, selected gene polymorphisms of IL-2R, IL-4, IL-6, IL-13 and IL-22 were identified as a possible risk factor related to MS development. Treatment strategies of multiple sclerosis that either target or utilize these cytokines seem rather promising, but more comprehensive research is necessary to gain a clearer understanding of how these cytokines precisely affect MS development and progression.
Collapse
Affiliation(s)
- Cezary Grunwald
- Department of Neurology, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A, 15-276 Białystok, Poland; (J.K.); (A.K.)
| | - Anna Krętowska-Grunwald
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, Jerzego Waszyngtona 17, 15-274 Białystok, Poland;
| | - Edyta Adamska-Patruno
- Clinical Research Center, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A, 15-276 Białystok, Poland;
| | - Jan Kochanowicz
- Department of Neurology, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A, 15-276 Białystok, Poland; (J.K.); (A.K.)
| | - Alina Kułakowska
- Department of Neurology, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A, 15-276 Białystok, Poland; (J.K.); (A.K.)
| | - Monika Chorąży
- Department of Neurology, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A, 15-276 Białystok, Poland; (J.K.); (A.K.)
| |
Collapse
|
5
|
Rodriguez-Mogeda C, van Ansenwoude CMJ, van der Molen L, Strijbis EMM, Mebius RE, de Vries HE. The role of CD56 bright NK cells in neurodegenerative disorders. J Neuroinflammation 2024; 21:48. [PMID: 38350967 PMCID: PMC10865604 DOI: 10.1186/s12974-024-03040-8] [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: 11/10/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
Emerging evidence suggests a potential role for natural killer (NK) cells in neurodegenerative diseases, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. However, the precise function of NK cells in these diseases remains ambiguous. The existence of two NK cell subsets, CD56bright and CD56dim NK cells, complicates the understanding of the contribution of NK cells in neurodegeneration as their functions within the context of neurodegenerative diseases may differ significantly. CD56bright NK cells are potent cytokine secretors and are considered more immunoregulatory and less terminally differentiated than their mostly cytotoxic CD56dim counterparts. Hence, this review focusses on NK cells, specifically on CD56bright NK cells, and their role in neurodegenerative diseases. Moreover, it explores the mechanisms underlying their ability to enter the central nervous system. By consolidating current knowledge, we aim to provide a comprehensive overview on the role of CD56bright NK cells in neurodegenerative diseases. Elucidating their impact on neurodegeneration may have implications for future therapeutic interventions, potentially ameliorating disease pathogenesis.
Collapse
Affiliation(s)
- Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Chaja M J van Ansenwoude
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Lennart van der Molen
- IQ Health Science Department, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eva M M Strijbis
- Amsterdam Neuroscience, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Department of Neurology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Ning Z, Liu Y, Guo D, Lin WJ, Tang Y. Natural killer cells in the central nervous system. Cell Commun Signal 2023; 21:341. [PMID: 38031097 PMCID: PMC10685650 DOI: 10.1186/s12964-023-01324-9] [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: 07/18/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK) cells are essential components of the innate lymphoid cell family that work as both cytotoxic effectors and immune regulators. Accumulating evidence points to interactions between NK cells and the central nervous system (CNS). Here, we review the basic knowledge of NK cell biology and recent advances in their roles in the healthy CNS and pathological conditions, with a focus on normal aging, CNS autoimmune diseases, neurodegenerative diseases, cerebrovascular diseases, and CNS infections. We highlight the crosstalk between NK cells and diverse cell types in the CNS and the potential value of NK cells as novel therapeutic targets for CNS diseases. Video Abstract.
Collapse
Affiliation(s)
- Zhiyuan Ning
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ying Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Daji Guo
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
7
|
Peng Y, Tao Y, Zhang Y, Wang J, Yang J, Wang Y. CD25: A potential tumor therapeutic target. Int J Cancer 2023; 152:1290-1303. [PMID: 36082452 DOI: 10.1002/ijc.34281] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 02/03/2023]
Abstract
CD25 is the alpha-chain of the heterotrimer IL-2 receptor. CD25 is expressed on the surface of both immune and non-immune cells with different frequencies. For cancers, CD25 is expressed at high levels in many types of hematological malignancies, but at low levels in most solid tumors. CD25 is also highly expressed in activated circulating immune cells and regulatory T cells (Tregs). Infiltration of Tregs in the tumor microenvironment can lead to an imbalanced ratio of effector T cells (Teffs) and Tregs, which is associated with the progression of cancers. A rescued Teff/Treg cell ratio indicates an efficient anti-tumor response to immunotherapy. CD25 as a potential target for the depletion of Tregs is critical in developing new immunotherapeutic strategies. Few articles have summarized the relationships between CD25 and tumors, or the recent progress of drugs targeting CD25. In this paper, we will discuss the structures of IL-2 and IL-2R, the biological function of CD25 and its important role in tumor therapy. In addition, the latest research on drugs targeting CD25 has been summarized, providing guidance for future drug development.
Collapse
Affiliation(s)
- Yujia Peng
- State Key Laboratory of Biotherapy, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease- related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiran Tao
- State Key Laboratory of Biotherapy, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease- related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ya Zhang
- State Key Laboratory of Biotherapy, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease- related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jinliang Yang
- State Key Laboratory of Biotherapy, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease- related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease- related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Zhang S, Zhang J, Zhang X, Lv P, Guo S. The protective effect of total glucosides of white paeony capsules on experimental autoimmune encephalomyelitis. Immunobiology 2023; 228:152313. [PMID: 36586141 DOI: 10.1016/j.imbio.2022.152313] [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: 01/10/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
AIMS To learn about the effect and mechanism of total glucosides of white peony capsule (TGP), on experimental autoimmune encephalomyelitis (EAE), an acknowledged animal model of multiple sclerosis (MS). METHODS The rat model of EAE was induced by subcutaneous injection with guinea pig spinal cord homogenate. The severity of the disease model was assessed by clinical score, hematoxylin and eosin (H&E) and luxol fast blue (LFB). Immunohistochemical assay was used to observe the types of inflammatory cells and adhesive molecule expression. Enzyme-linked immunosorbent assay (ELISA) was applied to detect content of the stem cell growth factor / mast cell growth factor (scf/MGF), interleukin-6 (IL-6) and IL-2. Immunofluorescence assay was applied to observe the expression of connexin43 (Cx43), glial fibrillary acidic protein (GFAP), connexin47 (Cx47) and the monoclonal antibody anti-adenomatous polyposis coli (APC) clone CC1. RESULTS Compare with the animals in EAE model group, TGP treated rats (particularly those treated with high doses) showed a significant decrease in morbidity, clinical scores, CNS infiltration of inflammatory cells (including mononuclear macrophages, CD4+ and CD8+ T cells) and demyelination. The key adhesion molecule ICAM-1, cytokines IL-2、IL-6 and scf/MGF were significantly decreased with TGP treatment. Oppositely, PD-1, connexin47 in oligodendrocytes and connexin43 in astrocytes were elevated with TGP treatment. CONCLUSION To sum up, TGP exhibited a significantly prevention and treatment effect on EAE rat model, and this improvement was achieved through a combination way composed of glial and inflammatory cells, junction proteins, various factors including adhesion factors, interleukins and scf/MGF.
Collapse
Affiliation(s)
- Suzhi Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Jun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Peng Lv
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Sanxing Guo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
9
|
Kardas G, Panek M, Kuna P, Damiański P, Kupczyk M. Monoclonal antibodies in the management of asthma: Dead ends, current status and future perspectives. Front Immunol 2022; 13:983852. [PMID: 36561741 PMCID: PMC9763885 DOI: 10.3389/fimmu.2022.983852] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
Patients with moderate-to-severe asthma may now be treated using a variety of monoclonal antibodies that target key inflammatory cytokines involved in disease pathogenesis. Existing clinical data on anti-IgE, anti-IL-5 and other immunological pathways indicate these therapies to offer reduced exacerbation rates, improved lung function, greater asthma control and better quality of life. However, as several patients still do not achieve satisfactory clinical response with the antibodies available, many more biologics, aiming different immunological pathways, are under evaluation. This review summarizes recent data on existing and potential monoclonal antibodies in asthma. Recent advances have resulted in the registration of a new antibody targeting TSLP (tezepelumab), with others being under development. Some of the researched monoclonal antibodies (e.g. anti-IL-13 tralokinumab and lebrikizumab or anti-IL-17A secukinumab) have shown optimistic results in preliminary research; however, these have been discontinued in asthma clinical research. In addition, as available monoclonal antibody treatments have shown little benefit among patients with T2-low asthma, research continues in this area, with several antibodies in development. This article summarizes the available pre-clinical and clinical data on new and emerging drugs for treating severe asthma, discusses discontinued treatments and outlines future directions in this area.
Collapse
|
10
|
Zhu H, Li G, Yin J, Zhang H, Da Y, Li L. Anlotinib attenuates experimental autoimmune encephalomyelitis mice model of multiple sclerosis via modulating the differentiation of Th17 and Treg cells. Immunopharmacol Immunotoxicol 2022; 44:594-602. [PMID: 35638564 DOI: 10.1080/08923973.2022.2071722] [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: 11/05/2022]
Abstract
BACKGROUND In multiple sclerosis (MS), the imbalance between T helper (Th)-17 cells and regulatory T (Treg) cells are critical in autoimmune central nervous system (CNS) inflammation and demyelination. Experimental autoimmune encephalomyelitis (EAE) is an established mouse MS model and simulates MS at diverse levels. OBJECTIVES This study aims at investigating the impact of anlotinib on the clinical severity of EAE and CD4+ T cell differentiation. MATERIALS AND METHODS EAE-induced mice were treated with water (control) or 6 mg/kg anlotinib by gavage daily. At the peak of EAE, histopathological examination and flow cytometry analysis of CNS-infiltrating CD4+ T cells were performed. In vitro differentiation of CD4+ T cells under different conditions was detected by flow cytometry and quantitative real-time PCR. Finally, the impacts of anlotinib on the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the transcription levels of key genes involved in Th17 and Treg differentiation were tested. RESULTS Anlotinib attenuated the clinical severity of EAE and changed the frequencies of CNS-infiltrating CD4+ T cell subsets. Anlotinib inhibited the differentiation of Th17 cells in vitro, decreased the phosphorylation of STAT3, and reduced the expression of Rorc. Anlotinib promoted the differentiation of Treg cells and upregulated the expression levels of CD39 and CD73. DISCUSSION AND CONCLUSIONS Anlotinib alleviated the symptoms of EAE via inhibiting the Th17 cell differentiation and promoting Treg cell differentiation. Our study provides new opportunities for the exploitation of anlotinib as a therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Haoran Zhu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Guangliang Li
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Jie Yin
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Hong Zhang
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Yurong Da
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Long Li
- Department of Immunology, Tianjin Medical University, Tianjin, China.,Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin, China.,Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
11
|
Castillo-Salazar M, Sánchez-Muñoz F, Springall del Villar R, Navarrete-Vázquez G, Hernández-DiazCouder A, Mojica-Cardoso C, García-Jiménez S, Toledano-Jaimes C, Bernal-Fernández G. Nitazoxanide Exerts Immunomodulatory Effects on Peripheral Blood Mononuclear Cells from Type 2 Diabetes Patients. Biomolecules 2021; 11:1817. [PMID: 34944461 PMCID: PMC8699442 DOI: 10.3390/biom11121817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a low-grade inflammatory condition with abnormalities in the immune response mediated by T lymphocytes and macrophages. Drug repositioning for immunomodulatory molecules is an attractive proposal for treating T2D. Nitazoxanide (NTZ) is a broad-spectrum drug with promising immunomodulatory effects. Thus, we investigated the immunomodulatory effect of NTZ on peripheral blood mononuclear cells (PBMCs) from patients with T2D. METHODS Fifty patients with T2D were selected, and the proliferative response of T lymphocytes and the M1/M2 ratio of macrophages post cell culture were evaluated by flow cytometry, as well as measuring the concentration of cytokines by ELISA and the relative expression of microRNAs (miRNAs) related to the immune response by real-time PCR. RESULTS NTZ exerts an inhibitory effect on the cell proliferation of T lymphocytes stimulated with anti-CD3 and anti-CD28 antibodies without modifying cell viability, and significant decreases in the supernatant concentrations of interleukin (IL)-1β, IL-2, IL-6, IL-10, and IL-12. Furthermore, NTZ negatively regulates the relative expression of miR-155-5p without changes in miR-146a-5p. The M1/M2 ratio of monocytes/macrophages decreased the M1 and increased the M2 subpopulation by NTZ. CONCLUSIONS Our results suggest that NTZ exerts immunomodulatory effects on PBMCs from T2D patients, and shows potential alternative therapeutic benefits.
Collapse
Affiliation(s)
- Mauricio Castillo-Salazar
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Fausto Sánchez-Muñoz
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | - Rashidi Springall del Villar
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | - Gabriel Navarrete-Vázquez
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Adrián Hernández-DiazCouder
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | | | - Sara García-Jiménez
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Cairo Toledano-Jaimes
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Germán Bernal-Fernández
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| |
Collapse
|
12
|
Wang C, Lv L, Wu Q, Wang Z, Luo Z, Sui B, Zhou M, Fu ZF, Zhao L. The role of interferon regulatory factor 7 in the pathogenicity and immunogenicity of rabies virus in a mouse model. J Gen Virol 2021; 102. [PMID: 34661517 DOI: 10.1099/jgv.0.001665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rabies is a zoonotic disease caused by the rabies virus (RABV). RABV can lead to fatal encephalitis and is still a serious threat in most parts of the world. Interferon regulatory factor 7 (IRF7) is the main transcriptional regulator of type I IFN, and it is crucial for the induction of IFNα/β and the type I IFN-dependent immune response. In this study, we focused on the role of IRF7 in the pathogenicity and immunogenicity of RABV using an IRF7-/- mouse model. The results showed that the absence of IRF7 made mice more susceptible to RABV, because IRF7 restricted the replication of RABV in the early stage of infection. IRF7 deficiency affected the recruitment of plasmacytoid dendritic cells to the draining lymph nodes (dLNs), reduced the production of type I IFN and expression of IFN-stimulated genes. Furthermore, we found that the ability to produce specific RABV-neutralizing antibody was impaired in IRF7-/- mice. Consistently, IRF7 deficiency affected the recruitment of germinal-centre B cells to dLNs, and the generation of plasma cells and RABV-specific antibody secreting cells. Moreover, the absence of IRF7 downregulated the induction of IFN-γ and reduced type 1 T helper cell (Th1)-dependent antibody production. Collectively, our findings demonstrate that IRF7 promotes humoral immune responses and compromises the pathogenicity of RABV in a mouse model.
Collapse
Affiliation(s)
- Caiqian Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Lei Lv
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Qiong Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Zhaochen Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Baokun Sui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| |
Collapse
|
13
|
Therapeutic potential of the target on NLRP3 inflammasome in multiple sclerosis. Pharmacol Ther 2021; 227:107880. [PMID: 33901504 DOI: 10.1016/j.pharmthera.2021.107880] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Inflammasomes are multi-protein macromolecular complexes that typically comprise of three units, a sensor, an adaptor and procaspase-1. The assembly of each inflammasome is dictated by a unique pattern recognition receptors (PRRs) in response to pathogen-associated molecular patterns (PAMPs) or other endogenous danger-associated molecular patterns (DAMPs) in the cytosol of the host cells, and promote the maturation and secretion of IL-1β and IL-18 during the inflammatory process. Specific inflammasomes are involved in the host defense response against different pathogens, and the latter have evolved multiple corresponding mechanisms to inhibit inflammasome activation. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome is the best understood in terms of molecular mechanisms, and is a promising therapeutic target in immune-related disorders. Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination of white matter in the central nervous system, increased levels of IL-1β in the cerebrospinal fluid (CSF) of relapsed patients, and deposition of caspase-1 in the spinal cord. The direct involvement of the NLRP3 inflammasome in the occurrence and development of MS was ascertained in the experimental autoimmune encephalomyelitis (EAE) animal model. In this review, we have focused on the mechanisms underlying activation of the NLRP3 inflammasome in MS or EAE, as well as inhibitors that specifically target the complex and alleviate disease progression, in order to unearth new therapeutic strategies against MS.
Collapse
|
14
|
Gklinos P, Papadopoulou M, Stanulovic V, Mitsikostas DD, Papadopoulos D. Monoclonal Antibodies as Neurological Therapeutics. Pharmaceuticals (Basel) 2021; 14:ph14020092. [PMID: 33530460 PMCID: PMC7912592 DOI: 10.3390/ph14020092] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/08/2023] Open
Abstract
Over the last 30 years the role of monoclonal antibodies in therapeutics has increased enormously, revolutionizing treatment in most medical specialties, including neurology. Monoclonal antibodies are key therapeutic agents for several neurological conditions with diverse pathophysiological mechanisms, including multiple sclerosis, migraines and neuromuscular disease. In addition, a great number of monoclonal antibodies against several targets are being investigated for many more neurological diseases, which reflects our advances in understanding the pathogenesis of these diseases. Untangling the molecular mechanisms of disease allows monoclonal antibodies to block disease pathways accurately and efficiently with exceptional target specificity, minimizing non-specific effects. On the other hand, accumulating experience shows that monoclonal antibodies may carry class-specific and target-associated risks. This article provides an overview of different types of monoclonal antibodies and their characteristics and reviews monoclonal antibodies currently in use or under development for neurological disease.
Collapse
Affiliation(s)
- Panagiotis Gklinos
- Department of Neurology, KAT General Hospital of Attica, 14561 Athens, Greece;
| | - Miranta Papadopoulou
- Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Vid Stanulovic
- Global Pharmacovigilance, R&D Sanofi, 91385 Chilly-Mazarin, France;
| | - Dimos D. Mitsikostas
- 1st Neurology Department, Aeginition Hospital, National and Kapodistrian University of Athens, 11521 Athens, Greece;
| | - Dimitrios Papadopoulos
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 129 Vasilissis Sophias Avenue, 11521 Athens, Greece
- Salpetriere Neuropsychiatric Clinic, 149 Papandreou Street, Metamorphosi, 14452 Athens, Greece
- Correspondence:
| |
Collapse
|
15
|
Mimpen M, Muris AH, Rolf L, Gerlach O, Kuhle J, Hupperts R, Smolders J, Damoiseaux J. Prognostic value of natural killer cell/T cell ratios for disease activity in multiple sclerosis. Eur J Neurol 2020; 28:901-909. [PMID: 33326677 PMCID: PMC7898592 DOI: 10.1111/ene.14680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 01/01/2023]
Abstract
Background and purpose Natural killer (NK) cells may play a role in multiple sclerosis (MS). Ratios of NK cells to CD4+ T cells have been proposed as a biomarker for the therapeutic effect of stem cell transplantation in MS. The objectives here were to explore the relevance of this ratio in MS patients by analysing NK and T cell subsets, as well as their prognostic value for disease activity. Methods Baseline peripheral blood mononuclear cells of 50 relapsing–remitting MS patients, participating in our vitamin D supplementation study (SOLARIUM), were analysed with flow cytometry. Disease activity was measured as new magnetic resonance imaging lesions, relapses and mean plasma neurofilament light chain levels after 48 weeks of follow‐up. Results The proportion of NK cells correlated negatively with CD4+ T cells (R = −0.335, p = 0.001) and interleukin 17A (IL‐17A+) CD4+ T cells (R = −0.203, p = 0.043). Participants with magnetic resonance imaging activity or relapses displayed lower NK/IL‐17A+ CD4+ T cell ratios (p =0.025 and p = 0.006, respectively). The NK/IL‐17A+ CD4+ T cell ratio correlated negatively with neurofilament light chain levels (R = −0.320, p = 0.050). Vitamin D supplementation did not affect these ratios. Conclusions Our data suggest a protective role of an expanded NK cell compartment compared to the CD4+ T cell subset fractions in relapsing–remitting MS patients. NK/CD4+ T cell ratios may be a prognostic biomarker for disease activity in MS.
Collapse
Affiliation(s)
- Max Mimpen
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Anne-Hilde Muris
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Linda Rolf
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Oliver Gerlach
- Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
| | - Jens Kuhle
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Raymond Hupperts
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Zuyderland Medical Center, Sittard, The Netherlands
| | - Joost Smolders
- MS Center ErasMS, Departments of Neurology and Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
16
|
Mayer P. Modelling bioactivities of combinations of whole extracts of edibles with a simplified theoretical framework reveals the statistical role of molecular diversity and system complexity in their mode of action and their nearly certain safety. PLoS One 2020; 15:e0239841. [PMID: 32986750 PMCID: PMC7521709 DOI: 10.1371/journal.pone.0239841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/14/2020] [Indexed: 11/25/2022] Open
Abstract
Network pharmacology and polypharmacology are emerging as novel drug discovery paradigms. The many discovery, safety and regulatory issues they raise may become tractable with polypharmacological combinations of natural compounds found in whole extracts of edible and mixes thereof. The primary goal of this work is to get general insights underlying the innocuity and the emergence of beneficial and toxic activities of combinations of many compounds in general and of edibles in particular. A simplified model of compounds’ interactions with an organism and of their desired and undesired effects is constructed by considering the departure from equilibrium of interconnected biological features. This model allows to compute the scaling of the probability of significant effects relative to nutritional diversity, organism complexity and synergy resulting from mixing compounds and edibles. It allows also to characterize massive indirect perturbation mode of action drugs as a potential novel multi-compound-multi-target pharmaceutical class, coined Ediceuticals when based on edibles. Their mode of action may readily target differentially organisms’ system robustness as such based on differential complexity for discovering nearly certainly safe novel antimicrobials, antiviral and anti-cancer treatments. This very general model provides also a theoretical framework to several pharmaceutical and nutritional observations. In particular, it characterizes two classes of undesirable effects of drugs, and may question the interpretation of undesirable effects in healthy subjects. It also formalizes nutritional diversity as such as a novel statistical supra-chemical parameter that may contribute to guide nutritional health intervention. Finally, it is to be noted that a similar formalism may be further applicable to model whole ecosystems in general.
Collapse
|
17
|
Ranganath T, Simpson LJ, Ferreira AM, Seiler C, Vendrame E, Zhao N, Fontenot JD, Holmes S, Blish CA. Characterization of the Impact of Daclizumab Beta on Circulating Natural Killer Cells by Mass Cytometry. Front Immunol 2020; 11:714. [PMID: 32391016 PMCID: PMC7194113 DOI: 10.3389/fimmu.2020.00714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Daclizumab beta is a humanized monoclonal antibody that binds to CD25 and selectively inhibits high-affinity IL-2 receptor signaling. As a former treatment for relapsing forms of multiple sclerosis (RMS), daclizumab beta induces robust expansion of the CD56bright subpopulation of NK cells that is correlated with the drug’s therapeutic effects. As NK cells represent a heterogeneous population of lymphocytes with a range of phenotypes and functions, the goal of this study was to better understand how daclizumab beta altered the NK cell repertoire to provide further insight into the possible mechanism(s) of action in RMS. We used mass cytometry to evaluate expression patterns of NK cell markers and provide a comprehensive assessment of the NK cell repertoire in individuals with RMS treated with daclizumab beta or placebo over the course of 1 year. Treatment with daclizumab beta significantly altered the NK cell repertoire compared to placebo treatment. As previously reported, daclizumab beta significantly increased expression of CD56 on total NK cells. Within the CD56bright NK cells, treatment was associated with multiple phenotypic changes, including increased expression of NKG2A and NKp44, and diminished expression of CD244, CD57, and NKp46. These alterations occurred broadly across the CD56bright population, and were not associated with a specific subset of CD56bright NK cells. While the changes were less dramatic, CD56dim NK cells responded distinctly to daclizumab beta treatment, with higher expression of CD2 and NKG2A, and lower expression of FAS-L, HLA-DR, NTB-A, NKp30, and Perforin. Together, these data indicate that the expanded CD56bright NK cells share features of both immature and mature NK cells. These findings show that daclizumab beta treatment is associated with unique changes in NK cells that may enhance their ability to kill autoreactive T cells or to exert immunomodulatory functions.
Collapse
Affiliation(s)
- Thanmayi Ranganath
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Laura J Simpson
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Anne-Maud Ferreira
- Department of Statistics, Stanford University, Stanford, CA, United States
| | - Christof Seiler
- Department of Statistics, Stanford University, Stanford, CA, United States
| | - Elena Vendrame
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Nancy Zhao
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | | | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, CA, United States
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
18
|
Mimpen M, Smolders J, Hupperts R, Damoiseaux J. Natural killer cells in multiple sclerosis: A review. Immunol Lett 2020; 222:1-11. [PMID: 32113900 DOI: 10.1016/j.imlet.2020.02.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
As the most common non-traumatic disabling disease among adolescents, multiple sclerosis (MS) is a devastating neurological inflammatory disease of the central nervous system. Research has not yet fully elucidated its pathogenesis, but it has shown MS to be a complex, multifactorial disease with many interplaying factors. One of these factors, natural killer (NK) cells, lymphocytes of the innate immune system, have recently gained attention due to the effects of daclizumab therapy, causing an expansion of the immunoregulatory subset of NK cells. Since then, NK cells and their relation to MS have been the focus of research, with many new findings being published in the last decade. In this review, NK cells are pictured as potent cytotoxic killers, as well as unique immune-regulators. Additionally, an overview of our current knowledge regarding NK cells in MS is given. The role of NK cells in MS is reviewed in the context of well-established environmental factors and current disease modifying therapies to gain further understanding of the pathogenesis and treatment options in MS.
Collapse
Affiliation(s)
- Max Mimpen
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands
| | - Joost Smolders
- Department of Neurology, Erasmus University Medical Center, Rotterdam The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam The Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands; Department of Neurology, Zuyderland Medical Center, Sittard The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht The Netherlands.
| |
Collapse
|
19
|
Ziliotto N, Marchetti G, Scapoli C, Bovolenta M, Meneghetti S, Benazzo A, Lunghi B, Balestra D, Laino LA, Bozzini N, Guidi I, Salvi F, Straudi S, Gemmati D, Menegatti E, Zamboni P, Bernardi F. C6orf10 Low-Frequency and Rare Variants in Italian Multiple Sclerosis Patients. Front Genet 2019; 10:573. [PMID: 31297130 PMCID: PMC6607989 DOI: 10.3389/fgene.2019.00573] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
In light of the complex nature of multiple sclerosis (MS) and the recently estimated contribution of low-frequency variants into disease, decoding its genetic risk components requires novel variant prioritization strategies. We selected, by reviewing MS Genome Wide Association Studies (GWAS), 107 candidate loci marked by intragenic single nucleotide polymorphisms (SNPs) with a remarkable association (p-value ≤ 5 × 10-6). A whole exome sequencing (WES)-based pilot study of SNPs with minor allele frequency (MAF) ≤ 0.04, conducted in three Italian families, revealed 15 exonic low-frequency SNPs with affected parent-child transmission. These variants were detected in 65/120 Italian unrelated MS patients, also in combination (22 patients). Compared with databases (controls gnomAD, dbSNP150, ExAC, Tuscany-1000 Genome), the allelic frequencies of C6orf10 rs16870005 and IL2RA rs12722600 were significantly higher (i.e., controls gnomAD, p = 9.89 × 10-7 and p < 1 × 10-20). TET2 rs61744960 and TRAF3 rs138943371 frequencies were also significantly higher, except in Tuscany-1000 Genome. Interestingly, the association of C6orf10 rs16870005 (Ala431Thr) with MS did not depend on its linkage disequilibrium with the HLA-DRB1 locus. Sequencing in the MS cohort of the C6orf10 3′ region revealed 14 rare mutations (10 not previously reported). Four variants were null, and significantly more frequent than in the databases. Further, the C6orf10 rare variants were observed in combinations, both intra-locus and with other low-frequency SNPs. The C6orf10 Ser389Xfr was found homozygous in a patient with early onset of the MS. Taking into account the potentially functional impact of the identified exonic variants, their expression in combination at the protein level could provide functional insights in the heterogeneous pathogenetic mechanisms contributing to MS.
Collapse
Affiliation(s)
- Nicole Ziliotto
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giovanna Marchetti
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Chiara Scapoli
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Matteo Bovolenta
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Silvia Meneghetti
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Andrea Benazzo
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Barbara Lunghi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Dario Balestra
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Lorenza Anna Laino
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Nicolò Bozzini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Irene Guidi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Fabrizio Salvi
- IRCCS Institute of Neurological Sciences, Hospital Bellaria, Bologna, Italy
| | - Sofia Straudi
- Department of Neurosciences and Rehabilitation, S. Anna University Hospital, Ferrara, Italy
| | - Donato Gemmati
- Department of Biomedical & Specialty Surgical Sciences and Centre Haemostasis & Thrombosis, Section of Medical Biochemistry, Molecular Biology & Genetics, University of Ferrara, Ferrara, Italy
| | - Erica Menegatti
- Department of Morphology, Surgery and Experimental Medicine, Vascular Diseases Center, University of Ferrara, Ferrara, Italy
| | - Paolo Zamboni
- Department of Morphology, Surgery and Experimental Medicine, Vascular Diseases Center, University of Ferrara, Ferrara, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|