98%
921
2 minutes
20
Background: Immune cell metabolism governs the outcome of immune responses and contributes to the development of autoimmunity by controlling lymphocyte pathogenic potential. In this study, we evaluated the metabolic profile of myelin-specific murine encephalitogenic T cells, to identify novel therapeutic targets for autoimmune neuroinflammation.
Methods: We performed metabolomics analysis on actively-proliferating encephalitogenic T cells to study their overall metabolic profile in comparison to resting T cells. Metabolomics, phosphoproteomics, in vitro functional assays, and in vivo studies in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), were then implemented to evaluate the effect of metabolic targeting on autoreactive T cell pathogenicity. Finally, we confirmed the translational potential of our targeting approach in human pro-inflammatory T helper cell subsets and in T cells from MS patients.
Results: We found that autoreactive encephalitogenic T cells display an altered coenzyme A (CoA) synthesis pathway, compared to resting T cells. CoA fueling with the CoA precursor pantethine (PTTH) affected essential immune-related processes of myelin-specific T cells, such as cell proliferation, cytokine production, and cell adhesion, both in vitro and in vivo. Accordingly, pre-clinical treatment with PTTH before disease onset inhibited the development of EAE by limiting T cell pro-inflammatory potential in vivo. Importantly, PTTH also significantly ameliorated the disease course when administered after disease onset in a therapeutic setting. Finally, PTTH reduced pro-inflammatory cytokine production by human T helper 1 (Th1) and Th17 cells and by T cells from MS patients, confirming its translational potential.
Conclusion: Our data demonstrate that CoA fueling with PTTH in pro-inflammatory and autoreactive T cells may represent a novel therapeutic approach for the treatment of autoimmune neuroinflammation.
Download full-text PDF |
Source |
---|---|
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC11536535 | PMC |
http://dx.doi.org/10.1186/s12974-024-03270-w | DOI Listing |
Int Immunol
August 2025
Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan.
The role of the thyroid hormone receptor beta (TR-β) in the immune system remains poorly understood; although its effect on TGF-βsignaling has been reported in non-immune systems. Here, we report that Thrb is highly expressed in pathogenic CD4+ T cells that infiltrate the central nervous system during experimental autoimmune encephalomyelitis (EAE) and Thrb is exclusively expressed in IL-17-producing CD4+ T cells (Th17 cells) that develop both in vitro or in vivo. Sobetirome, a selective TR-βagonist, promoted pathogenic Th17 differentiation and IL-17 production in the presence of exogenous IL-1β.
View Article and Find Full Text PDFSci Transl Med
August 2025
Univ Toulouse, INSERM, CNRS, Infinity, Toulouse, France.
Follicular regulatory T cells (T cells) constitute a subset of regulatory T cells pivotal to the immune response in germinal centers (GCs) that inhibit autoantibody production. Their role, however, remains ill-defined in autoimmune diseases like multiple sclerosis (MS) and its murine model, experimental autoimmune encephalomyelitis (EAE), which are neuroinflammatory diseases driven by T and B cells. Here, we quantified peripheral blood immune subpopulations in two cohorts of patients with MS and found higher circulating T cell frequencies in patients in relapse compared with patients in remission.
View Article and Find Full Text PDFImmunity
September 2025
Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China. Electronic address:
B cells are central drivers of central nervous system (CNS) autoimmune disorders, including multiple sclerosis (MS). Although the brain meninges normally maintain a stringently non-self-reactive B cell repertoire, how disruption of this local immune tolerance contributes to pathology remains unclear. Here, we demonstrated that autoreactive B cells at the brain border accelerated neuroinflammation by directly engaging encephalitogenic T cells.
View Article and Find Full Text PDFbioRxiv
June 2025
Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.
Amino acids play critical roles in the activation and function of lymphocytes. Here we show that the non-essential amino acid, asparagine, is essential for optimal activation and proliferation of CD4 T cells. We demonstrate that asparagine depletion at different time points after CD4 T cell activation reduces mitochondrial membrane potential and function.
View Article and Find Full Text PDFNature
July 2025
Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA.
The microbiota has been recognized as a critical contributor to various diseases, with multiple reports of changes in the composition of the gut microbiome in contexts such as inflammatory bowel disease and neurodegenerative diseases. These microbial shifts can exert systemic effects by altering the release of specific metabolites into the bloodstream, and the gastrointestinal microbiota has also been reported to exhibit immunomodulatory activity through the activation of innate and adaptive immunity. However, it remains unclear how the microbiota contributes to inflammation in the central nervous system (CNS), where these microorganisms are typically absent.
View Article and Find Full Text PDF