1
|
Scherrer EC, Ramos KA, Valadares YM, Soares IGM, Carli AP, Sá Silva F, Silva JG, Alvarenga DG, Brugiolo ASS, Verly RM, Salvador MR, Denadai AML, Souza Alves CC, Castro SBR. Ursolic acid derivatives improved clinical signs of experimental autoimmune encephalomyelitis by modulating central nervous system inflammation. Metab Brain Dis 2025; 40:166. [PMID: 40167825 DOI: 10.1007/s11011-025-01591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
The immunopathogenesis of multiple sclerosis (MS) involves the activation of T lymphocytes, leading to progressive axonal loss and brain atrophy. Ursolic acid (AU) has been widely used as an herbal medicine, with the ability to inhibit the production and secretion of cytokines and may influence the differentiation of CD4 + helper cells. In this study, we aimed to investigate the immunomodulatory effects of ursolic acid derivatives (methyl 3β-hydroxyurs-12-en-28-oate (AUD1) and methyl 3β-acetoxyurs-12-en-28-oate (AUD2)) in a model of experimental autoimmune encephalomyelitis (EAE). EAE was induced by subcutaneous immunization of myelin oligodendrocyte glycoprotein peptide (MOG35-55) in C57BL/6 mice. On the 15th day post-induction (dpi), the mice were treated with AU, AUD1, or AUD2 (50 mg/kg intraperitoneally per day) for six days. Clinical signs were monitored until 21 dpi, and parameters were assessed in the spinal cord, lymph nodes, and brain at 21 dpi. The results showed that both derivatives similarly attenuated the clinical signs of EAE and reduced inflammation and demyelination in the spinal cord. In addition, they reduced the number of pro-inflammatory cells in the brain, the level of IL-1β, TNF, and IFN-γ in the spinal cord, and, in the periphery, promoted the regulation of pro-inflammatory cells. In conclusion, regulating cells in the periphery and reducing the number of pro-inflammatory cells in the CNS, with AUD1 and AUD2, culminated in the efficacy of the clinical parameters presented in EAE, suggesting a therapeutic potential for treating MS.
Collapse
Affiliation(s)
- Elaine Carlos Scherrer
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Karla Antunes Ramos
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Ydia Mariele Valadares
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Igor Gabriel Machado Soares
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Alessandra Paula Carli
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Fernando Sá Silva
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Jeferson Gomes Silva
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Daniel Gomes Alvarenga
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | | | - Rodrigo Moreira Verly
- Department of Chemistry, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, MG, Brazil
| | | | | | - Caio César Souza Alves
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil.
- Faculdade de Medicina do Mucuri, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Rua do Cruzeiro, 01, Jardim São Paulo, Teófilo Otoni/MG, Brasil, 39803-371.
| | | |
Collapse
|
2
|
Ma X, Zhang J, Jiang Q, Li YX, Yang G. Human microbiome-derived peptide affects the development of experimental autoimmune encephalomyelitis via molecular mimicry. EBioMedicine 2025; 111:105516. [PMID: 39724786 PMCID: PMC11732510 DOI: 10.1016/j.ebiom.2024.105516] [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: 07/31/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Gut commensal microbiota has been identified as a potential environmental risk factor for multiple sclerosis (MS), and numerous studies have linked the commensal microorganism with the onset of MS. However, little is known about the mechanisms underlying the gut microbiome and host-immune system interaction. METHODS We employed bioinformatics methodologies to identify human microbial-derived peptides by analyzing their similarity to the MHC II-TCR binding patterns of self-antigens. Subsequently, we conducted a range of in vitro and in vivo assays to assess the encephalitogenic potential of these microbial-derived peptides. FINDINGS We analyzed 304,246 human microbiome genomes and 103 metagenomes collected from the MS cohort and identified 731 nonredundant analogs of myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). Of note, half of these analogs could bind to MHC II and interact with TCR through structural modeling of the interaction using fine-tuned AlphaFold. Among the 8 selected peptides, the peptide (P3) shows the ability to activate MOG35-55-specific CD4+ T cells in vitro. Furthermore, P3 shows encephalitogenic capacity and has the potential to induce EAE in some animals. Notably, mice immunized with a combination of P3 and MOG35-55 develop severe EAE. Additionally, dendritic cells could process and present P3 to MOG35-55-specific CD4+ T cells and activate these cells. INTERPRETATION Our data suggests the potential involvement of a MOG35-55-mimic peptide derived from the gut microbiota as a molecular trigger of EAE pathogenesis. Our findings offer direct evidence of how microbes can initiate the development of EAE, suggesting a potential explanation for the correlation between certain gut microorganisms and MS prevalence. FUNDING National Natural Science Foundation of China (82371350 to GY).
Collapse
MESH Headings
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Animals
- Molecular Mimicry
- Mice
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/chemistry
- Gastrointestinal Microbiome
- Peptides/chemistry
- Peptides/immunology
- Peptide Fragments/immunology
- Peptide Fragments/chemistry
- Disease Models, Animal
- Receptors, Antigen, T-Cell/metabolism
- Computational Biology/methods
- Histocompatibility Antigens Class II/metabolism
- Protein Binding
- Microbiota
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Multiple Sclerosis
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
Collapse
Affiliation(s)
- Xin Ma
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Jian Zhang
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Qianling Jiang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yong-Xin Li
- Department of Chemistry and the Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China; Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
3
|
Fan H, Fu Q, Du G, Qin L, Shi X, Wang D, Yang Y. Microglial Mayhem NLRP3 Inflammasome's Role in Multiple Sclerosis Pathology. CNS Neurosci Ther 2024; 30:e70135. [PMID: 39690733 DOI: 10.1111/cns.70135] [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: 12/19/2023] [Revised: 09/29/2024] [Accepted: 11/05/2024] [Indexed: 12/19/2024] Open
Abstract
INTRODUCTION This review delves into the intricate relationship between NLR inflammasomes, particularly the NLRP3 inflammasome, and the immune-mediated neurodegenerative disease, multiple sclerosis (MS). While the precise etiology of MS remains elusive, compelling research underscores the pivotal role of the immune response in disease progression. Notably, recent investigations highlight the significant involvement of NLRP3 inflammasomes in various autoimmune diseases, prompting an in-depth exploration of their impact on MS. METHOD The review focuses on elucidating the activation mechanism of NLRP3 inflammasomes within microglia/macrophages (MG/MФ), examining how this activation promotes an inflammatory response that exacerbates neuronal damage in MS. A comprehensive analysis of existing literature and research findings forms the basis for understanding the intricate interplay between NLRP3 inflammasomes and MS pathogenesis. RESULTS Synthesizing current research, the review provides insight into the pivotal role played by NLR inflammasomes, specifically NLRP3, in MS. Emphasis is placed on the inflammatory response orchestrated by activated MG/MФ, elucidating the cascade that perpetuates neuronal damage in the disease. CONCLUSIONS This review concludes by consolidating key findings and offering a nuanced perspective on the role of NLRP3 inflammasomes in MS pathogenesis. The detailed exploration of the activation process within MG/MФ provides a foundation for understanding the disease's underlying mechanisms. Furthermore, the review sets the stage for potential therapeutic strategies targeting NLRP3 inflammasomes in the pursuit of MS treatment.
Collapse
Affiliation(s)
- Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Qizhi Fu
- Department of Intensive Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Dongmei Wang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Yanhui Yang
- Department of Emergency Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
4
|
Morandi E, Adoue V, Bernard I, Friebel E, Nunez N, Aubert Y, Masson F, Dejean AS, Becher B, Astier A, Martinet L, Saoudi A. Impact of the Multiple Sclerosis-Associated Genetic Variant CD226 Gly307Ser on Human CD8 T-Cell Functions. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200306. [PMID: 39231385 PMCID: PMC11379124 DOI: 10.1212/nxi.0000000000200306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/08/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND AND OBJECTIVES The rs763361 nonsynonymous variant in the CD226 gene, which results in a glycine-to-serine substitution at position 307 of the CD226 protein, has been implicated as a risk factor of various immune-mediated diseases, including multiple sclerosis (MS). Compelling evidence suggests that this allele may play a significant role in predisposing individuals to MS by decreasing the immune-regulatory capacity of Treg cells and increasing the proinflammatory potential of effector CD4 T cells. However, the impact of this CD226 gene variant on CD8 T-cell functions, a population that also plays a key role in MS, remains to be determined. METHODS To study whether the CD226 risk variant affects human CD8 T-cell functions, we used CD8 T cells isolated from peripheral blood mononuclear cell of 16 age-matched healthy donors homozygous for either the protective or the risk allele of CD226. We characterized these CD8 T cells on T-cell receptor (TCR) stimulation using high-parametric flow cytometry and bulk RNAseq and through characterization of canonical signaling pathways and cytokine production. RESULTS On TCR engagement, the phenotype of ex vivo CD8 T cells bearing the protective (CD226-307Gly) or the risk (CD226-307Ser) allele of CD226 was largely overlapping. However, the transcriptomic signature of CD8 T cells from the donors carrying the risk allele presented an enrichment in TCR, JAK/STAT, and IFNγ signaling. We next found that the CD226-307Ser risk allele leads to a selective increase in the phosphorylation of the mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 (ERK1/2) associated with enhanced phosphorylation of STAT4 and increased production of IFNγ. DISCUSSION Our data suggest that the CD226-307Ser risk variant imposes immune dysregulation by increasing the pathways related to IFNγ signaling in CD8 T cells, thereby contributing to the risk of developing chronic inflammation.
Collapse
Affiliation(s)
- Elena Morandi
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Véronique Adoue
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Isabelle Bernard
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Ekaterina Friebel
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Nicolas Nunez
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Yann Aubert
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Frederick Masson
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Anne S Dejean
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Burkhard Becher
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Anne Astier
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Ludovic Martinet
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| | - Abdelhadi Saoudi
- From the Infinity-Toulouse Institute for Infectious and Inflammatory Diseases (E.M., V.A., I.B., Y.A., F.M., A.S.D., A.A., A.S.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1291, Centre National de la Recherche Scientifique (CNRS) UMR 5051, Université Paul Sabatier (UPS), Toulouse, France; Institute of Experimental Immunology (E.F., N.N., B.B.), University of Zurich, Switzerland; and Cancer Research Center of Toulouse (CRCT) (L.M.), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
5
|
Kneuer JM, Grajek IA, Winkler M, Erbe S, Meinecke T, Weiss R, Garfias-Veitl T, Sheikh BN, König AC, Möbius-Winkler MN, Kogel A, Kresoja KP, Rosch S, Kokot KE, Filipova V, Gaul S, Thiele H, Lurz P, von Haehling S, Speer T, Laufs U, Boeckel JN. Novel Long Noncoding RNA HEAT4 Affects Monocyte Subtypes, Reducing Inflammation and Promoting Vascular Healing. Circulation 2024; 150:1101-1120. [PMID: 39005211 PMCID: PMC11444369 DOI: 10.1161/circulationaha.124.069315] [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] [Accepted: 06/11/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Activation of the immune system contributes to cardiovascular diseases. The role of human-specific long noncoding RNAs in cardioimmunology is poorly understood. METHODS Single-cell sequencing in peripheral blood mononuclear cells revealed a novel human-specific long noncoding RNA called HEAT4 (heart failure-associated transcript 4). HEAT4 expression was assessed in several in vitro and ex vivo models of immune cell activation, as well as in the blood of patients with heart failure (HF), acute myocardial infarction, or cardiogenic shock. The transcriptional regulation of HEAT4 was verified through cytokine treatment and single-cell sequencing. Loss-of-function and gain-of-function studies and multiple RNA-protein interaction assays uncovered a mechanistic role of HEAT4 in the monocyte anti-inflammatory gene program. HEAT4 expression and function was characterized in a vascular injury model in NOD.CB17-Prkdc scid/Rj mice. RESULTS HEAT4 expression was increased in the blood of patients with HF, acute myocardial infarction, or cardiogenic shock. HEAT4 levels distinguished patients with HF from people without HF and predicted all-cause mortality in a cohort of patients with HF over 7 years of follow-up. Monocytes, particularly anti-inflammatory CD16+ monocytes, which are increased in patients with HF, are the primary source of HEAT4 expression in the blood. HEAT4 is transcriptionally activated by treatment with anti-inflammatory interleukin-10. HEAT4 activates anti-inflammatory and inhibits proinflammatory gene expression. Increased HEAT4 levels result in a shift toward more CD16+ monocytes. HEAT4 binds to S100A9, causing a monocyte subtype switch, thereby reducing inflammation. As a result, HEAT4 improves endothelial barrier integrity during inflammation and promotes vascular healing after injury in mice. CONCLUSIONS These results characterize a novel endogenous anti-inflammatory pathway that involves the conversion of monocyte subtypes into anti-inflammatory CD16+ monocytes. The data identify a novel function for the class of long noncoding RNAs by preventing protein secretion and suggest long noncoding RNAs as potential targets for interventions in the field of cardioimmunology.
Collapse
Affiliation(s)
- Jasmin M. Kneuer
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Ignacy A. Grajek
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Melanie Winkler
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Stephan Erbe
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Tim Meinecke
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Ronald Weiss
- Institute of Clinical Immunology, University of Leipzig, Germany (R.W.)
| | - Tania Garfias-Veitl
- Department of Cardiology and Pneumology, University Medical Center of Göttingen (UMG), Germany (T.G.-V., S.v.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany (T.G.-V., S.v.H.)
| | - Bilal N. Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Germany (B.N.S.)
| | - Ann-Christine König
- German Research Center for Environmental Health (GmbH), Metabolomics and Proteomics Core, Helmholtz Zentrum München, Germany (A.-C.K.)
| | - Maximilian N. Möbius-Winkler
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Alexander Kogel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Karl-Patrik Kresoja
- Department of Cardiology, Heart Center at University of Leipzig, Germany (K.-P.K., S.R., H.T., P.L.)
- Department of Cardiology, Universitätsmedizin Johannes Gutenberg-University, Mainz, Germany (K.-P.K., S.R., P.L.)
| | - Sebastian Rosch
- Department of Cardiology, Heart Center at University of Leipzig, Germany (K.-P.K., S.R., H.T., P.L.)
- Department of Cardiology, Universitätsmedizin Johannes Gutenberg-University, Mainz, Germany (K.-P.K., S.R., P.L.)
| | - Karoline E. Kokot
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Vanina Filipova
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Susanne Gaul
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Holger Thiele
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
- Department of Cardiology, Heart Center at University of Leipzig, Germany (K.-P.K., S.R., H.T., P.L.)
| | - Philipp Lurz
- Department of Cardiology, Heart Center at University of Leipzig, Germany (K.-P.K., S.R., H.T., P.L.)
- Department of Cardiology, Universitätsmedizin Johannes Gutenberg-University, Mainz, Germany (K.-P.K., S.R., P.L.)
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medical Center of Göttingen (UMG), Germany (T.G.-V., S.v.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany (T.G.-V., S.v.H.)
| | - Thimoteus Speer
- Medizinische Klinik 4: Nephrologie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany (T.S.)
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University, Frankfurt, Germany (T.S.)
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Germany (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., U.L., J.-N.B.)
- Central German Heart Alliance (J.M.K., I.A.G., M.W., S.E., T.M., M.N.M.-W., A.K., K.E.K., V.F., S.G., H.T., U.L., J.-N.B.)
| |
Collapse
|
6
|
Yang C, Ma Y, Lu Q, Qu Y, Li Y, Cheng S, Xiao C, Chen J, Wang C, Wang F, Xiang AP, Huang W, Tang X, Zheng H. 2-Bromo-1,4-Naphthalenedione promotes CD8 + T cell expansion and limits Th1/Th17 to mitigate experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:181. [PMID: 39068463 PMCID: PMC11283727 DOI: 10.1186/s12974-024-03172-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: 04/24/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Treating Multiple sclerosis (MS), a well-known immune-mediated disease characterized by axonal demyelination, is challenging due to its complex causes. Naphthalenedione, present in numerous plants, is being explored as a potential medicine for MS due to its immunomodulatory properties. However, its effects on lymphocytes can vary depending on factors such as the specific compound, concentration, and experimental conditions. In this study, we aim to explore the therapeutic potential of 2-bromo-1,4-naphthalenedione (BrQ), a derivative of naphthalenedione, in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and to elucidate its underlying mechanisms. We observed that mice treated with BrQ exhibited reduced severity of EAE symptoms, including lower clinical scores, decreased leukocyte infiltration, and less extensive demyelination in central nervous system. Furthermore, it was noted that BrQ does not directly affect the remyelination process. Through cell-chat analysis based on bulk RNA-seq data, coupled with validation of flow analysis, we discovered that BrQ significantly promotes the expansion of CD8+ T cells and their interactions with other immune cells in peripheral immune system in EAE mice. Subsequent CD8+ T cell depletion experiments confirmed that BrQ alleviates EAE in a CD8+ T cell-dependent manner. Mechanistically, expanded CD8+ cells were found to selectively reduce antigen-specific CD4+ cells and subsequently inhibit Th1 and Th17 cell development in vivo, ultimately leading to relief from EAE. In summary, our findings highlight the crucial role of BrQ in modulating the pathogenesis of MS, suggesting its potential as a novel drug candidate for treating MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Cuixia Yang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuanchen Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiying Lu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuliang Qu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yuantao Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chongjun Xiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chuangjia Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Feng Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China.
| | - Xiaorong Tang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China.
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
7
|
Hai-Na Z, Jun-Jie J, Guang-Meng X. Peptides derived from growth factors: Exploring their diverse impact from antimicrobial properties to neuroprotection. Biomed Pharmacother 2024; 176:116830. [PMID: 38824833 DOI: 10.1016/j.biopha.2024.116830] [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: 01/29/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Growth factor-derived peptides are bioactive molecules that play a crucial role in various physiological processes within the human body. Over the years, extensive research has revealed their diverse applications, ranging from antimicrobial properties to their potential in neuroprotection and treating various diseases. These peptides exhibit innate immune responses and have been found to possess potent antimicrobial properties against a wide range of pathogens. Growth factor-derived peptides have demonstrated the ability to promote neuronal survival, prevent cell death, and stimulate neural regeneration. As a result, they hold immense promise in the treatment of various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis, as well as in the management of traumatic brain injuries. Moreover, growth factor-derived peptides have shown potential for supporting tissue repair and wound healing processes. By enhancing cell proliferation and migration, these peptides contribute to the regeneration of damaged tissues and promote a more efficient healing response. The applications of growth factor-derived peptides extend beyond their therapeutic potential in health; they also have a role in various disease conditions. For example, researchers have explored their influence on cancer cells, where some peptides have demonstrated anti-cancer properties, inhibiting tumor growth and promoting apoptosis in cancer cells. Additionally, their immunomodulatory properties have been investigated for potential applications in autoimmune disorders. Despite the immense promise shown by growth factor-derived peptides, some challenges need to be addressed. Nevertheless, ongoing research and advancements in biotechnology offer promising avenues to overcome these obstacles. The review summarizes the foundational biology of growth factors and the intricate signaling pathways in various physiological processes as well as diseases such as cancer, neurodegenerative disorders, cardiovascular ailments, and metabolic syndromes.
Collapse
Affiliation(s)
- Zhang Hai-Na
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130000, PR China
| | - Jiang Jun-Jie
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130000, PR China
| | - Xu Guang-Meng
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun 130000, PR China.
| |
Collapse
|
8
|
Sharon N, Yarmolinsky L, Khalfin B, Fleisher-Berkovich S, Ben-Shabat S. Cannabinoids' Role in Modulating Central and Peripheral Immunity in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:6402. [PMID: 38928109 PMCID: PMC11204381 DOI: 10.3390/ijms25126402] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Cannabinoids (the endocannabinoids, the synthetic cannabinoids, and the phytocannabinoids) are well known for their various pharmacological properties, including neuroprotective and anti-inflammatory features, which are fundamentally important for the treatment of neurodegenerative diseases. The aging of the global population is causing an increase in these diseases that require the development of effective drugs to be even more urgent. Taking into account the unavailability of effective drugs for neurodegenerative diseases, it seems appropriate to consider the role of cannabinoids in the treatment of these diseases. To our knowledge, few reviews are devoted to cannabinoids' impact on modulating central and peripheral immunity in neurodegenerative diseases. The objective of this review is to provide the best possible information about the cannabinoid receptors and immuno-modulation features, peripheral immune modulation by cannabinoids, cannabinoid-based therapies for the treatment of neurological disorders, and the future development prospects of making cannabinoids versatile tools in the pursuit of effective drugs.
Collapse
Affiliation(s)
| | | | | | | | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (N.S.); (L.Y.); (B.K.); (S.F.-B.)
| |
Collapse
|
9
|
Ruiz-Fernández I, Sánchez-Díaz R, Ortega-Sollero E, Martín P. Update on the role of T cells in cognitive impairment. Br J Pharmacol 2024; 181:799-815. [PMID: 37559406 DOI: 10.1111/bph.16214] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/03/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
The central nervous system (CNS) has long been considered an immune-privileged site, with minimal interaction between immune cells, particularly of the adaptive immune system. Previously, the presence of immune cells in this organ was primarily linked to events involving disruption of the blood-brain barrier (BBB) or inflammation. However, current research has shown that immune cells are found patrolling CNS under homeostatic conditions. Specifically, T cells of the adaptive immune system are able to cross the BBB and are associated with ageing and cognitive impairment. In addition, T-cell infiltration has been observed in pathological conditions, where inflammation correlates with poor prognosis. Despite ongoing research, the role of this population in the ageing brain under both physiological and pathological conditions is not yet fully understood. In this review, we provide an overview of the interactions between T cells and other immune and CNS parenchymal cells, and examine the molecular mechanisms by which these interactions may contribute to normal brain function and the scenarios in which disruption of these connections lead to cognitive impairment. A comprehensive understanding of the role of T cells in the ageing brain and the underlying molecular pathways under normal conditions could pave the way for new research to better understand brain disorders. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
| | - Raquel Sánchez-Díaz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | | | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
10
|
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
|
11
|
Bisht P, Rathore C, Rathee A, Kabra A. Astrocyte Activation and Drug Target in Pathophysiology of Multiple Sclerosis. Methods Mol Biol 2024; 2761:431-455. [PMID: 38427254 DOI: 10.1007/978-1-0716-3662-6_30] [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: 03/02/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease, which is also referred to as an autoimmune disorder with chronic inflammatory demyelination affecting the core system that is the central nervous system (CNS). Demyelination is a pathological manifestation of MS. It is the destruction of myelin sheath, which is wrapped around the axons, and it results in the loss of synaptic connections and conduction along the axon is also compromised. Various attempts are made to understand MS and demyelination using various experimental models out of them. The most popular model is experimental autoimmune encephalomyelitis (EAE), in which autoimmunity against CNS components is induced in experimental animals by immunization with self-antigens derived from basic myelin protein. Astrocytes serve as a dual-edged sword both in demyelination and remyelination. Various drug targets have also been discussed that can be further explored for the treatment of MS. An extensive literature research was done from various online scholarly and research articles available on PubMed, Google Scholar, and Elsevier. Keywords used for these articles were astrocyte, demyelination, astrogliosis, and reactive astrocytes. This includes articles being the most relevant information to the area compiled to compose a current review.
Collapse
Affiliation(s)
- Preeti Bisht
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Charul Rathore
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Ankit Rathee
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Ajitgarh, Punjab, India
| |
Collapse
|
12
|
Tanaka H, Hasebe R, Murakami K, Sugawara T, Yamasaki T, Murakami M. Gateway reflexes describe novel neuro-immune communications that establish immune cell gateways at specific vessels. Bioelectron Med 2023; 9:24. [PMID: 37936169 PMCID: PMC10631009 DOI: 10.1186/s42234-023-00126-1] [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: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Neuroinflammation is an important biological process induced by complex interactions between immune cells and neuronal cells in the central nervous system (CNS). Recent research on the bidirectional communication between neuronal and immunological systems has provided evidence for how immune and inflammatory processes are regulated by nerve activation. One example is the gateway reflex, in which immune cells bypass the blood brain barrier and infiltrate the CNS to cause neuroinflammation. We have found several modes of the gateway reflex in mouse models, in which gateways for immune cells are established at specific blood vessels in the spinal cords and brain in experimental autoimmune encephalomyelitis and systemic lupus erythematosus models, at retinal blood vessels in an experimental autoimmune uveitis model, and the ankle joints in an inflammatory arthritis model. Several environmental stimulations, including physical and psychological stresses, activate neurological pathways that alter immunological responses via the gateway reflex, thus contributing to the development/suppression of autoimmune diseases. In the manuscript, we describe the discovery of the gateway reflex and recent insights on how they regulate disease development. We hypothesize that artificial manipulation of specific neural pathways can establish and/or close the gateways to control the development of autoimmune diseases.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
| | - Rie Hasebe
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Kaoru Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Toshiki Sugawara
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
| | - Takeshi Yamasaki
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-Ku, Sapporo, 060-0815, Japan.
- Division of Molecular Neuroimmunology, National Institute for Physiological Sciences, national Institute for Natural Sciences, Nishi-38, Myodaiji-cho, Okazaki, 444-8585, Japan.
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Anagawa 4-9-1, Inage-Ku, Chiba, 263-8555, Japan.
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Nishi-11, Kita-21, Kuta-Ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
13
|
de Fàbregues O, Sellés M, Ramos-Vicente D, Roch G, Vila M, Bové J. Relevance of tissue-resident memory CD8 T cells in the onset of Parkinson's disease and examination of its possible etiologies: infectious or autoimmune? Neurobiol Dis 2023; 187:106308. [PMID: 37741513 DOI: 10.1016/j.nbd.2023.106308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/05/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023] Open
Abstract
Tissue-resident memory CD8 T cells are responsible for local immune surveillance in different tissues, including the brain. They constitute the first line of defense against pathogens and cancer cells and play a role in autoimmunity. A recently published study demonstrated that CD8 T cells with markers of residency containing distinct granzymes and interferon-γ infiltrate the parenchyma of the substantia nigra and contact dopaminergic neurons in an early premotor stage of Parkinson's disease. This infiltration precedes α-synuclein aggregation and neuronal loss in the substantia nigra, suggesting a relevant role for CD8 T cells in the onset of the disease. To date, the nature of the antigen that initiates the adaptive immune response remains unknown. This review will discuss the role of tissue-resident memory CD8 T cells in brain immune homeostasis and in the onset of Parkinson's disease and other neurological diseases. We also discuss how aging and genetic factors can affect the CD8 T cell immune response and how animal models can be misleading when studying human-related immune response. Finally, we speculate about a possible infectious or autoimmune origin of Parkinson's disease.
Collapse
Affiliation(s)
- Oriol de Fàbregues
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Department, Vall d'Hebron University Hospital
| | - Maria Sellés
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - David Ramos-Vicente
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Gerard Roch
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Catalonia, Spain; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Jordi Bové
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain.
| |
Collapse
|
14
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
15
|
Ellen O, Ye S, Nheu D, Dass M, Pagnin M, Ozturk E, Theotokis P, Grigoriadis N, Petratos S. The Heterogeneous Multiple Sclerosis Lesion: How Can We Assess and Modify a Degenerating Lesion? Int J Mol Sci 2023; 24:11112. [PMID: 37446290 DOI: 10.3390/ijms241311112] [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: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous disease of the central nervous system that is governed by neural tissue loss and dystrophy during its progressive phase, with complex reactive pathological cellular changes. The immune-mediated mechanisms that promulgate the demyelinating lesions during relapses of acute episodes are not characteristic of chronic lesions during progressive MS. This has limited our capacity to target the disease effectively as it evolves within the central nervous system white and gray matter, thereby leaving neurologists without effective options to manage individuals as they transition to a secondary progressive phase. The current review highlights the molecular and cellular sequelae that have been identified as cooperating with and/or contributing to neurodegeneration that characterizes individuals with progressive forms of MS. We emphasize the need for appropriate monitoring via known and novel molecular and imaging biomarkers that can accurately detect and predict progression for the purposes of newly designed clinical trials that can demonstrate the efficacy of neuroprotection and potentially neurorepair. To achieve neurorepair, we focus on the modifications required in the reactive cellular and extracellular milieu in order to enable endogenous cell growth as well as transplanted cells that can integrate and/or renew the degenerative MS plaque.
Collapse
Affiliation(s)
- Olivia Ellen
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Danica Nheu
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| |
Collapse
|
16
|
Packer D, Fresenko EE, Harrington EP. Remyelination in animal models of multiple sclerosis: finding the elusive grail of regeneration. Front Mol Neurosci 2023; 16:1207007. [PMID: 37448959 PMCID: PMC10338073 DOI: 10.3389/fnmol.2023.1207007] [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: 04/16/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Remyelination biology and the therapeutic potential of restoring myelin sheaths to prevent neurodegeneration and disability in multiple sclerosis (MS) has made considerable gains over the past decade with many regeneration strategies undergoing tested in MS clinical trials. Animal models used to investigate oligodendroglial responses and regeneration of myelin vary considerably in the mechanism of demyelination, involvement of inflammatory cells, neurodegeneration and capacity for remyelination. The investigation of remyelination in the context of aging and an inflammatory environment are of considerable interest for the potential translation to progressive multiple sclerosis. Here we review how remyelination is assessed in mouse models of demyelination, differences and advantages of these models, therapeutic strategies that have emerged and current pro-remyelination clinical trials.
Collapse
|
17
|
Thevarkalam M, Krishnan S, Shanmughan LI, Mathai A, Leelamani JV, Kannoth S, Bhaskaran R, Iype T, Panda S. Determination of sensitivities and specificities of cerebrospinal fluid free light chains to diagnose multiple sclerosis- a multicentric case-control study. Mult Scler Relat Disord 2023; 74:104717. [PMID: 37062197 DOI: 10.1016/j.msard.2023.104717] [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: 08/10/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND CSF free light chains help diagnose multiple sclerosis, but no data is available on the Asian population. Our objective was to study the diagnostic utility of CSF free light chains for diagnosing multiple sclerosis in Indian patients. METHODS Prospective multicentric case-control study. Cases included those who were tested for oligoclonal bands and fulfilled the modified McDonald criteria 2017 for multiple sclerosis and clinically isolated syndromes. Those tested for oligoclonal bands (OCB) but with other diagnoses- inflammatory and non-inflammatory were included as controls. Clinical details were collected from electronic medical records. CSF and serum kappa and lambda free light chains were measured, apart from oligoclonal bands, immunoglobulin, and albumin in paired serum and CSF samples. RESULTS There were 70 patients (31 cases and 39 controls). The mean age was 43.41(SD 16.073) years, and 43(61.4%) were females. CSF kappa showed highest specificity 97.4%, at a cut off 2.06 mg/L (sensitivity 71%) and highest sensitivity 90.3%, at a cut off 0.47 mg/L (specificity 79.5%). Best balance of sensitivity and specificity for CSF kappa was seen at a cut-off of ≥ 0.63 mg/L {sensitivity 87·1 (CI - 70.17-96.37), and specificity 87·18 (CI -72.57-95.70)}. The ratio of Kappa/lambda showed highest specificity of 100%(similar to OCB) with a sensitivity of 71% at a cut off of 1.72. The ratio of sum of kappa and lambda light chains, and Qalb (∑CSF FLC/Qalb), showed the highest specificity (94.87%)among the blood brain barrier corrected ratios. CONCLUSION This study showed that the diagnostic utility of CSF kappa was comparable to OCB to diagnose multiple sclerosis in sensitivity, but not specificity, so can be a screening test before testing for OCB in our population.
Collapse
Affiliation(s)
- Meena Thevarkalam
- Department of Biochemistry, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Sajitha Krishnan
- Department of Biochemistry, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041.
| | - Layana I Shanmughan
- Department of Biochemistry, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Annamma Mathai
- Neuroimmunology Laboratory, Department of Neurology, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Jyothi V Leelamani
- Neuroimmunology Laboratory, Department of Neurology, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Sudheeran Kannoth
- Neuroimmunology Laboratory, Department of Neurology, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Renjitha Bhaskaran
- Department of biostatistics, Amrita Institute of Medical Sciences, Amrita Viswavidyapeetham University, Kochi, Kerala, India 682041
| | - Thomas Iype
- Department of Neurology, Government Medical College, Thiruvananthapuram, Kerala, India. 695011
| | - Samhita Panda
- Department of Neurology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India 342005
| |
Collapse
|
18
|
Sardari E, Ebadi A, Razzaghi-Asl N. In silico repurposing of CNS drugs for multiple sclerosis. Mult Scler Relat Disord 2023; 73:104622. [PMID: 36958175 DOI: 10.1016/j.msard.2023.104622] [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: 03/28/2022] [Revised: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune neurodegenerative disease affecting numerous people worldwide. While the relapsing subtypes of MS are to some extent treatable, the disease remains incurable leading to progressive disability. Limited efficacy of current small molecule drugs necessitates development of efficient and safe MS medications. Accordingly, drug repurposing is an invaluable strategy that recognizes new targets for known drugs especially in the field of poorly addressed therapeutic areas. Drug discovery largely depends on the identification of potential binding molecules to the intended biomolecular target(s). In this regard, current study was devoted to in silico repurposing of 263 small molecule CNS drugs to achieve superior binders to some MS-related targets. On the basis of molecular docking scores, thioxanthene and benzisothiazole-based antipsychotics could be identified as potential binders to sphingosine-1-phosphate lyase (S1PL) and cyclophilin D (CypD). Tightest interaction modes were observed for zuclopenthixol-S1PL (ΔGb -7.96 kcal/mol) and lurasidone-CypD (ΔGb -8.84 kcal/mol) complexes. Molecular dynamics (MD) simulations proved the appropriate and stable accommodation of top-ranked drugs inside enzyme binding sites during 100 ns. Hydroxyethyl piperazine of zuclopenthixol and benzisothiazole of lurasidone flipped inside the binding pocket to interact with adjacent polar and apolar residues. Solvent accessible surface area (SASA) fluctuations confirmed the results of binding trajectory analysis and showed that non-polar hydrophobic interactions played significant roles in acquired stabilities. Our results on lurasidone binding pattern were interestingly in accordance with previous reports on X-ray structures of other norbornane maleimide derivatives as CypD inhibitors. According to this, Asn144, Phe102 and Phe155 served as important residues in providing stable binding pose of lurasidone through both exo and endo conformations. Although experimental results are necessary to be achieved, the outcomes of this study proposed the potentiality of some thioxanthene and benzisothiazole-based antipsychotics for binding to S1PL and CypD, respectively, as MS-related targets.
Collapse
Affiliation(s)
- Elham Sardari
- Student Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ahmad Ebadi
- Department of Medicinal Chemistry, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nima Razzaghi-Asl
- Department of Medicinal Chemistry School of Pharmacy, Ardabil University of Medical Sciences, Ardabil PO code: 5618953141, Iran.
| |
Collapse
|
19
|
Innate and adaptive immune abnormalities underlying autoimmune diseases: the genetic connections. SCIENCE CHINA. LIFE SCIENCES 2023:10.1007/s11427-021-2187-3. [PMID: 36738430 PMCID: PMC9898710 DOI: 10.1007/s11427-021-2187-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023]
Abstract
With the exception of an extremely small number of cases caused by single gene mutations, most autoimmune diseases result from the complex interplay between environmental and genetic factors. In a nutshell, etiology of the common autoimmune disorders is unknown in spite of progress elucidating certain effector cells and molecules responsible for pathologies associated with inflammatory and tissue damage. In recent years, population genetics approaches have greatly enriched our knowledge regarding genetic susceptibility of autoimmunity, providing us with a window of opportunities to comprehensively re-examine autoimmunity-associated genes and possible pathways. In this review, we aim to discuss etiology and pathogenesis of common autoimmune disorders from the perspective of human genetics. An overview of the genetic basis of autoimmunity is followed by 3 chapters detailing susceptibility genes involved in innate immunity, adaptive immunity and inflammatory cell death processes respectively. With such attempts, we hope to expand the scope of thinking and bring attention to lesser appreciated molecules and pathways as important contributors of autoimmunity beyond the 'usual suspects' of a limited subset of validated therapeutic targets.
Collapse
|
20
|
Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nat Rev Immunol 2022; 22:734-750. [PMID: 35508809 DOI: 10.1038/s41577-022-00718-z] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Our incomplete understanding of the causes and pathways involved in the onset and progression of multiple sclerosis (MS) limits our ability to effectively treat this complex neurological disease. Recent studies explore the role of immune cells at different stages of MS and how they interact with cells of the central nervous system (CNS). The findings presented here begin to question the exclusivity of an antigen-specific cause and highlight how seemingly distinct immune cell types can share common functions that drive disease. Innovative techniques further expose new disease-associated immune cell populations and reinforce how environmental context is critical to their phenotype and subsequent role in disease. Importantly, the differentiation of immune cells into a pathogenic state is potentially reversible through therapeutic manipulation. As such, understanding the mechanisms that provide plasticity to causal cell types is likely key to uncoupling these disease processes and may identify novel therapeutic targets that replace the need for cell ablation.
Collapse
Affiliation(s)
- Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
21
|
Fournier AP, Tastet O, Charabati M, Hoornaert C, Bourbonnière L, Klement W, Larouche S, Tea F, Wang YC, Larochelle C, Arbour N, Ragoussis J, Zandee S, Prat A. Single-Cell Transcriptomics Identifies Brain Endothelium Inflammatory Networks in Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 10:10/1/e200046. [PMID: 36446612 PMCID: PMC9709715 DOI: 10.1212/nxi.0000000000200046] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/31/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND AND OBJECTIVES Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease characterized by infiltration of immune cells in multifocal areas of the CNS. The specific molecular processes allowing autoreactive immune cells to enter the CNS compartment through the blood-brain barrier remain elusive. METHODS Using endothelial cell (EC) enrichment and single-cell RNA sequencing, we characterized the cells implicated in the neuroinflammatory processes in experimental autoimmune encephalomyelitis, an animal model of MS. Validations on human MS brain sections of the most differentially expressed genes in venous ECs were performed using immunohistochemistry and confocal microscopy. RESULTS We found an upregulation of genes associated with antigen presentation and interferon in most populations of CNS-resident cells, including ECs. Interestingly, instead of transcriptionally distinct profiles, a continuous gradient of gene expression separated the arteriovenous zonation of the brain vasculature. However, differential gene expression analysis presented more transcriptomic alterations on the venous side of the axis, suggesting a prominent role of venous ECs in neuroinflammation. Furthermore, analysis of ligand-receptor interactions identified important potential molecular communications between venous ECs and infiltrated immune populations. To confirm the relevance of our observation in the context of human disease, we validated the protein expression of the most upregulated genes (Ackr1 and Lcn2) in MS lesions. DISCUSSION In this study, we provide a landscape of the cellular heterogeneity associated with neuroinflammation. We also present important molecular insights for further exploration of specific cell processes that promote infiltration of immune cells inside the brain of experimental autoimmune encephalomyelitis mice.
Collapse
Affiliation(s)
- Antoine Philippe Fournier
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Olivier Tastet
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Marc Charabati
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Chloé Hoornaert
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Lyne Bourbonnière
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Wendy Klement
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Sandra Larouche
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Fiona Tea
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Yu Chang Wang
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Catherine Larochelle
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Nathalie Arbour
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Jiannis Ragoussis
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Stephanie Zandee
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada
| | - Alexandre Prat
- From the Neuroimmunology Research Laboratory (A.P.F., O.T., M.C., C.H., L.B., W.K., S.L., F.T., C.L., N.A., S.Z., A.P.), Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM); Department of Neurosciences (A.P.F., C.L., N.A., S.Z., A.P.), Faculty of Medicine, Université de Montréal; Multiple Sclerosis Clinic (C.L., A.P.), Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM); Department of Human Genetics (J.R.), McGill University, Montréal; and McGill Genome Centre (Y.C.W., J.R.), Montréal, Québec, Canada.
| |
Collapse
|
22
|
Bild W, Vasincu A, Rusu RN, Ababei DC, Stana AB, Stanciu GD, Savu B, Bild V. Impact of the Renin-Angiotensin System on the Pathogeny and Pharmacotherapeutics of Neurodegenerative Diseases. Biomolecules 2022; 12:1429. [PMID: 36291638 PMCID: PMC9599929 DOI: 10.3390/biom12101429] [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: 08/14/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
Brain neurodegenerative diseases (BND) are debilitating conditions that are especially characteristic of a certain period of life and considered major threats to human health. Current treatments are limited, meaning that there is a challenge in developing new options that can efficiently tackle the different components and pathophysiological processes of these conditions. The renin-angiotensin-aldosterone system (RAS) is an endocrine axis with important peripheral physiological functions such as blood pressure and cardiovascular homeostasis, as well as water and sodium balance and systemic vascular resistance-functions which are well-documented. However, recent work has highlighted the paracrine and autocrine functions of RAS in different tissues, including the central nervous system (CNS). It is known that RAS hyperactivation has pro-inflammatory and pro-oxidant effects, thus suggesting that its pharmacological modulation could be used in the management of these conditions. The present paper underlines the involvement of RAS and its components in the pathophysiology of BNDs such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), motor neuron disease (MND), and prion disease (PRD), as well as the identification of drugs and pharmacologically active substances that act upon RAS, which could alleviate their symptomatology or evolution, and thus, contribute to novel therapeutic approaches.
Collapse
Affiliation(s)
- Walther Bild
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
| | - Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Aurelian Bogdan Stana
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Savu
- Department of Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Veronica Bild
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
23
|
Jayaraman M, Dutta P, Krishnan S, Arora K, Sivakumar D, Raghavendran HRB. Emerging Promise of Phytochemicals in Ameliorating Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-124961. [PMID: 35786341 DOI: 10.2174/1871527321666220701153926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/09/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The field of medicine and synthetic drug development have advanced rapidly over the past few decades. However, research on alternative medicine such as phytochemicals cannot be ignored. The main reason for prominent curiosity about phytochemicals stems from the belief that usage of natural compounds is safer and has lesser detrimental side effects. OBJECTIVE The aim of the present review was to discuss in detail with several phytochemicals that have been studied or are being studied in the context of various neurological disorders including depression, Alzheimer's disease, Huntington's disease and even neuroinflammatory disorders such as encephalitis. METHODS The potential role of phytochemicals in the treatment or management of symptoms associated with neurological disorders have been included in this article. All data included in this paper has been pooled from various databases including Google Scholar, PubMed, Science Direct, Springer and Wiley Online Library. RESULTS Phytochemicals have been widely studied for their therapeutic properties associated with neurological disorders. Using various experimental techniques for both in vivo and in vitro experiments, studies have shown that phytochemicals do have antioxidant, anti-inflammatory and neuroprotective activities which play major roles in the treatment of neurological diseases. CONCLUSION Even though there has been compelling evidence of the therapeutic role of phytochemicals, further research is still required to evaluate the safety and efficacy of these medicines. Using previously published papers as foundation for additional research such as preclinical studies and clinical trials, phytochemicals can become a safer alternative to synthetic drugs for treating a spectrum of neurological diseases.
Collapse
Affiliation(s)
- Megala Jayaraman
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603203, Chennai, Tamil Nadu, India
| | - Parijat Dutta
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603203, Chennai, Tamil Nadu, India
| | - Sabari Krishnan
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603203, Chennai, Tamil Nadu, India
| | - Khyati Arora
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603203, Chennai, Tamil Nadu, India
| | - Diveyaa Sivakumar
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur-603203, Chennai, Tamil Nadu, India
- School of Dental Sciences, University Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Hanumanth Rao Balaji Raghavendran
- Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Central Research Facility, Porur, Chennai-600116, India
| |
Collapse
|
24
|
Liu Z, Niu X, Wang J. Naringenin as a natural immunomodulator against T cell-mediated autoimmune diseases: literature review and network-based pharmacology study. Crit Rev Food Sci Nutr 2022; 63:11026-11043. [PMID: 35776085 DOI: 10.1080/10408398.2022.2092054] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
T cells, especially CD4+ T helper (Th) cells, play a vital role in the pathogenesis of specific autoimmune diseases. Naringenin, a citrus flavonoid, exhibits anti-inflammatory, anti-oxidant, and antitumor properties, which have been verified in animal autoimmune disease models. However, naringenin's possible effects and molecular mechanisms in T cell-mediated autoimmune diseases are unclear. This review summarizes the findings of previous studies and predicts the target of naringenin in T cell-mediated autoimmune disorders such as multiple sclerosis, inflammatory bowel disease, and rheumatoid arthritis through network pharmacology analysis. We performed DAVID enrichment analysis, protein-protein interaction analysis, and molecular docking to predict the positive effect of naringenin on T cell-mediated autoimmune disorders. Sixteen common genes were screened, among which the core genes were PTGS2, ESR1, CAT, CASP3, MAPK1, and AKT1. The possible molecular mechanism relates to HIF-1, estrogen, TNF, and NF-κB signaling pathways. Our findings have significance for future naringenin treatment of T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Zejin Liu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Xinli Niu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Junpeng Wang
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| |
Collapse
|
25
|
Zeng X, Zheng M, Liu T, Bahabayi A, Song S, Alimu X, Kang R, Lu S, Song Y, Liu C. Cytotoxic T Lymphocytes Expressing GPR56 are Up-regulated in the Peripheral Blood of Patients with Active Rheumatoid Arthritis and Reflect Disease Progression. Immunol Invest 2022; 51:1804-1819. [PMID: 35404706 DOI: 10.1080/08820139.2022.2058403] [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
OBJECTIVE This study aims to elucidate the changes in the percentage of GPR56 and/or granzyme B (GZMB) positive cells in rheumatoid arthritis (RA) CD4 and CD8 T lymphocytes, and to explore their clinical value in diagnosing and reflecting the progression of RA. METHODS The percentages of GPR56 and/or GZMB positive cells were analyzed in peripheral blood (PB) and spleen T cells in a collagen-induced arthritis (CIA) model established in DBA/1 mice. The percentages of GPR56+ and/or GZMB+ cells were further analyzed in PBs from RA patients and healthy controls. Correlation analysis was performed between clinical indicators and GPR56+, GZMB+, and GPR56+ GZMB+ T cells. Receiver operating characteristic (ROC) curves were used to evaluate the value of GPR56 and GZMB in differentiating active and stable remitting RA. RESULTS GPR56+ levels were increased in CD4 and CD8 T cells in the PB of CIA mice. The percentages of GPR56+ and GZMB+ cells were increased in both CD4 and CD8 T cell subsets in patients with active RA. GPR56+, GZMB+, and GPR56+ GZMB+ cells were positively correlated with rheumatoid factor and DAS28. ROC analysis revealed that AUCs for GPR56+, GZMB+, and GPR56+ GZMB+ cell percentages to distinguish active RA from stable remission RA were 0.7106, 0.6941, 0.7024, with cut-off values of 16.35, 16.40, 14.80 in CD4 + T cells, and 0.8031, 0.8086, 0.8196 with cut-off values 60.25, 62.15, 40.15 in CD8 + T cells, respectively. CONCLUSIONS GPR56+ and/or GZMB+ T cells are up-regulated in patients with active RA and reflect their condition. The detection of GPR56 and GZMB is helpful for RA disease assessment.
Collapse
Affiliation(s)
- Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Mohan Zheng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Tianci Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Shi Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiayidan Alimu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Rui Kang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Songsong Lu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ying Song
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
26
|
Vasincu A, Rusu RN, Ababei DC, Larion M, Bild W, Stanciu GD, Solcan C, Bild V. Endocannabinoid Modulation in Neurodegenerative Diseases: In Pursuit of Certainty. BIOLOGY 2022; 11:biology11030440. [PMID: 35336814 PMCID: PMC8945712 DOI: 10.3390/biology11030440] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 01/13/2023]
Abstract
Simple Summary Neurodegenerative diseases represent an important cause of morbidity and mortality worldwide. Existing therapeutic options are limited and focus mostly on improving symptoms and reducing exacerbations. The endocannabinoid system is involved in the pathophysiology of such disorders, an idea which has been highlighted by recent scientific work. The current work focusses its attention on the importance and implications of this system and its synthetic and natural ligands in disorders such as Alzheimer’s, Parkinson’s, Huntington’s and multiple sclerosis. Abstract Neurodegenerative diseases are an increasing cause of global morbidity and mortality. They occur in the central nervous system (CNS) and lead to functional and mental impairment due to loss of neurons. Recent evidence highlights the link between neurodegenerative and inflammatory diseases of the CNS. These are typically associated with several neurological disorders. These diseases have fundamental differences regarding their underlying physiology and clinical manifestations, although there are aspects that overlap. The endocannabinoid system (ECS) is comprised of receptors (type-1 (CB1R) and type-2 (CB2R) cannabinoid-receptors, as well as transient receptor potential vanilloid 1 (TRPV1)), endogenous ligands and enzymes that synthesize and degrade endocannabinoids (ECBs). Recent studies revealed the involvement of the ECS in different pathological aspects of these neurodegenerative disorders. The present review will explore the roles of cannabinoid receptors (CBRs) and pharmacological agents that modulate CBRs or ECS activity with reference to Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Huntington’s Disease (HD) and multiple sclerosis (MS).
Collapse
Affiliation(s)
- Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
- Correspondence:
| | - Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
| | - Mădălina Larion
- Department of Anaesthesiology Intensive Therapy, Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 19 Croitorilor Street, 400162 Cluj-Napoca, Romania;
- Department of Anaesthetics, Midland Regional Hospital, Longford Road, Mullingar, N91 NA43 Co. Westmeath, Ireland
| | - Walther Bild
- Department of Physiology, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Carmen Solcan
- Preclinics Department, “Ion Ionescu de la Brad” University of Life Sciences, 8 M. Sadoveanu Alley, 700489 Iasi, Romania;
| | - Veronica Bild
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| |
Collapse
|
27
|
Kronsten VT, Tranah TH, Pariante C, Shawcross DL. Gut-derived systemic inflammation as a driver of depression in chronic liver disease. J Hepatol 2022; 76:665-680. [PMID: 34800610 DOI: 10.1016/j.jhep.2021.11.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023]
Abstract
Depression and chronic liver disease (CLD) are important causes of disability, morbidity and mortality worldwide and their prevalence continues to rise. The rate of depression in CLD is high compared to that of the general population and is comparable to the increased rates observed in other medical comorbidities and chronic inflammatory conditions. Notably, a comorbid diagnosis of depression has a detrimental effect on outcomes in cirrhosis. Systemic inflammation is pivotal in cirrhosis-associated immune dysfunction - a phenomenon present in advanced CLD (cirrhosis) and implicated in the development of complications, organ failure, disease progression, increased infection rates and poor outcome. The presence of systemic inflammation is also well-documented in a cohort of patients with depression; peripheral cytokine signals can result in neuroinflammation, behavioural change and depressive symptoms via neural mechanisms, cerebral endothelial cell and circumventricular organ signalling, and peripheral immune cell-to-brain signalling. Gut dysbiosis has been observed in both patients with cirrhosis and depression. It leads to intestinal barrier dysfunction resulting in increased bacterial translocation, in turn activating circulating immune cells, leading to cytokine production and systemic inflammation. A perturbed gut-liver-brain axis may therefore explain the high rates of depression in patients with cirrhosis. The underlying mechanisms explaining the critical relationship between depression and cirrhosis remain to be fully elucidated. Several other psychosocial and biological factors are likely to be involved, and therefore the cause is probably multifactorial. However, the role of the dysfunctional gut-liver-brain axis as a driver of gut-derived systemic inflammation requires further exploration and consideration as a target for the treatment of depression in patients with cirrhosis.
Collapse
Affiliation(s)
- Victoria T Kronsten
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK.
| | - Thomas H Tranah
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Carmine Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, The Maurice Wohl Clinical Neuroscience Institute, Cutcombe Road, London, SE5 9RT, UK
| | - Debbie L Shawcross
- Institute of Liver Studies, 1(st) Floor James Black Centre, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| |
Collapse
|
28
|
Song YC, Liu CT, Lee HJ, Yen HR. Cordycepin prevents and ameliorates experimental autoimmune encephalomyelitis by inhibiting leukocyte infiltration and reducing neuroinflammation. Biochem Pharmacol 2022; 197:114918. [DOI: 10.1016/j.bcp.2022.114918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022]
|
29
|
Pachner AR. The Neuroimmunology of Multiple Sclerosis: Fictions and Facts. Front Neurol 2022; 12:796378. [PMID: 35197914 PMCID: PMC8858985 DOI: 10.3389/fneur.2021.796378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
There have been tremendous advances in the neuroimmunology of multiple sclerosis over the past five decades, which have led to improved diagnosis and therapy in the clinic. However, further advances must take into account an understanding of some of the complex issues in the field, particularly an appreciation of "facts" and "fiction." Not surprisingly given the incredible complexity of both the nervous and immune systems, our understanding of the basic biology of the disease is very incomplete. This lack of understanding has led to many controversies in the field. This review identifies some of these controversies and facts/fictions with relation to the basic neuroimmunology of the disease (cells and molecules), and important clinical issues. Fortunately, the field is in a healthy transition from excessive reliance on animal models to a broader understanding of the disease in humans, which will likely lead to many improved treatments especially of the neurodegeneration in multiple sclerosis (MS).
Collapse
Affiliation(s)
- Andrew R. Pachner
- Dartmouth–Hitchcock Medical Center, Lebanon, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
30
|
Krajewski D, Paul D, Ge S, Jellison E, Pachter JS. Appearance of claudin-5 + leukocyte subtypes in the blood and CNS during progression of EAE. J Neuroinflammation 2021; 18:296. [PMID: 34933669 PMCID: PMC8691042 DOI: 10.1186/s12974-021-02328-3] [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: 06/08/2021] [Accepted: 11/23/2021] [Indexed: 11/24/2022] Open
Abstract
Background Tight junctions (TJs) are membrane specializations characteristic of barrier-forming membranes, which function to seal the aqueous pathway between endothelial cells or epithelial cells and, thereby, obstruct intercellular solute and cellular movement. However, previous work from our laboratory found that claudin-5 (CLN-5), a TJ protein prominent at the blood–brain barrier (BBB), was also detected, ectopically, on leukocytes (CLN-5+) in the blood and central nervous system (CNS) of mice with experimental autoimmune encephalomyelitis (EAE), a neuroinflammatory, demyelinating disease that is a model for multiple sclerosis. CLN-5 was further shown to be transferred from endothelial cells to circulating leukocytes during disease, prompting consideration this action is coupled to leukocyte transendothelial migration (TEM) into the CNS by fostering transient interactions between corresponding leukocyte and endothelial junctional proteins at the BBB. Methods To begin clarifying the significance of CLN-5+ leukocytes, flow cytometry was used to determine their appearance in the blood and CNS during EAE. Results Flow cytometric analysis revealed CLN-5+ populations among CD4 and CD8 T cells, B cells, monocytes and neutrophils, and these appeared with varying kinetics and to different extents in both blood and CNS. CLN-5 levels on circulating T cells further correlated highly with activation state. And, the percentage of CLN-5+ cells among each of the subtypes analyzed was considerably higher in CNS tissue than in blood, consistent with the interpretation that CLN-5+ leukocytes gain preferred access to the CNS. Conclusion Several leukocyte subtypes variably acquire CLN-5 in blood before they enter the CNS, an event that may represent a novel mechanism to guide leukocytes to sites for paracellular diapedesis across the BBB. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02328-3.
Collapse
Affiliation(s)
- Dylan Krajewski
- Blood-Brain Barrier Laboratory, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA.,Department of Immunology, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA.,Department of Immunology, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA.,PureTech Health, 6 Tide Street, Boston, MA, 02210, USA
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA.,Department of Immunology, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Evan Jellison
- Department of Immunology, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA. .,Department of Immunology, UConn Health, 263 Farmington Ave., Farmington, CT, 06030, USA.
| |
Collapse
|
31
|
Feizi N, Focaccetti C, Pacella I, Tucci G, Rossi A, Costanza M, Pedotti R, Sidney J, Sette A, La Rocca C, Procaccini C, Matarese G, Barnaba V, Piconese S. CD8 + T cells specific for cryptic apoptosis-associated epitopes exacerbate experimental autoimmune encephalomyelitis. Cell Death Dis 2021; 12:1026. [PMID: 34716313 PMCID: PMC8556378 DOI: 10.1038/s41419-021-04310-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/12/2021] [Accepted: 09/29/2021] [Indexed: 01/20/2023]
Abstract
The autoimmune immunopathology occurring in multiple sclerosis (MS) is sustained by myelin-specific and -nonspecific CD8+ T cells. We have previously shown that, in MS, activated T cells undergoing apoptosis induce a CD8+ T cell response directed against antigens that are unveiled during the apoptotic process, namely caspase-cleaved structural proteins such as non-muscle myosin and vimentin. Here, we have explored in vivo the development and the function of the immune responses to cryptic apoptosis-associated epitopes (AEs) in a well-established mouse model of MS, experimental autoimmune encephalomyelitis (EAE), through a combination of immunization approaches, multiparametric flow cytometry, and functional assays. First, we confirmed that this model recapitulated the main findings observed in MS patients, namely that apoptotic T cells and effector/memory AE-specific CD8+ T cells accumulate in the central nervous system of mice with EAE, positively correlating with disease severity. Interestingly, we found that AE-specific CD8+ T cells were present also in the lymphoid organs of unprimed mice, proliferated under peptide stimulation in vitro, but failed to respond to peptide immunization in vivo, suggesting a physiological control of this response. However, when mice were immunized with AEs along with EAE induction, AE-specific CD8+ T cells with an effector/memory phenotype accumulated in the central nervous system, and the disease severity was exacerbated. In conclusion, we demonstrate that AE-specific autoimmunity may contribute to immunopathology in neuroinflammation.
Collapse
Affiliation(s)
- Neda Feizi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Chiara Focaccetti
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy.,Department of Human Science and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166, Rome, Italy
| | - Ilenia Pacella
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Gloria Tucci
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Alessandra Rossi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy
| | - Massimo Costanza
- Molecular Neuro-Oncology Unit, Department of Clinical Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Rosetta Pedotti
- Molecular Neuro-Oncology Unit, Department of Clinical Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131, Naples, Italy
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131, Naples, Italy.,Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, 00143, Rome, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131, Naples, Italy
| | - Vincenzo Barnaba
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy. .,Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00161, Rome, Italy.
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161, Rome, Italy. .,Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, 00143, Rome, Italy. .,Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00161, Rome, Italy.
| |
Collapse
|
32
|
Kronsten VT, Shawcross DL. Hepatic encephalopathy and depression in chronic liver disease: is the common link systemic inflammation? Anal Biochem 2021; 636:114437. [PMID: 34715068 DOI: 10.1016/j.ab.2021.114437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/26/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023]
Abstract
Hepatic encephalopathy and depression share a number of clinical features, such as cognitive impairment and psychomotor retardation, and are highly prevalent in patients with chronic liver disease. Both conditions signify a poor prognosis, carry an increased mortality and are major determinants of reduced health related quality of life. The pathophysiology of hepatic encephalopathy is complex. Whilst cerebral accumulation of ammonia is well-recognised as being central to the development of hepatic encephalopathy, systemic inflammation, which acts in synergy with hyperammonaemia, is emerging as a key driver in its development. The pro-inflammatory state is also widely documented in depression, and peripheral to brain communication occurs resulting in central inflammation, behavioural changes and depressive symptoms. Gut dysbiosis, with a similar reduction in beneficial bacteria, increase in pathogens and decreased bacterial diversity, has been observed in both hepatic encephalopathy and depression, and it may be that the resultant increased bacterial translocation causes their shared inflammatory pathophysiology. Whilst the literature on a positive association between hepatic encephalopathy and depression in cirrhosis remains to be substantiated, there is evolving evidence that treatment with psychobiotics may be of dual benefit, improving cognition and mood in cirrhosis.
Collapse
Affiliation(s)
- Victoria Tatiana Kronsten
- Institute of Liver Studies, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, UK.
| | - Debbie Lindsay Shawcross
- Institute of Liver Studies, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, UK
| |
Collapse
|
33
|
Sutiwisesak R, Burns TC, Rodriguez M, Warrington AE. Remyelination therapies for multiple sclerosis: optimizing translation from animal models into clinical trials. Expert Opin Investig Drugs 2021; 30:857-876. [PMID: 34126015 DOI: 10.1080/13543784.2021.1942840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Multiple sclerosis (MS) is the most common inflammatory disease of the central nervous system (CNS). Demyelination, the main pathology in MS, contributes to clinical symptoms and long-term neurological deficits if left untreated. Remyelination, the natural repair of damaged myelin by cells of the oligodendrocyte lineage, occurs in MS, but eventually fails in most patients as they age. Encouraging timely remyelination can restore axon conduction and minimize deficits.Areas covered: We discuss and correlate human MS pathology with animal models, propose methods to deplete resident oligodendrocyte progenitor cells (OPCs) to determine whether mature oligodendrocytes support remyelination, and review remyelinating agents, mechanisms of action, and available clinical trial data.Expert opinion: The heterogeneity of human MS may limit successful translation of many candidate remyelinating agents; some patients lack the biological targets necessary to leverage current approaches. Development of therapeutics for remyelination has concentrated almost exclusively on mobilization of innate OPCs. However, mature oligodendrocytes appear an important contributor to remyelination in humans. Limiting the contribution of OPC mediated repair in models of MS would allow the evaluation of remyelination-promoting agents on mature oligodendrocytes. Among remyelinating reagents reviewed, only rHIgM22 targets both OPCs and mature oligodendrocytes.
Collapse
Affiliation(s)
- Rujapope Sutiwisesak
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Terry C Burns
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| | - Moses Rodriguez
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur E Warrington
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
34
|
Mockus TE, Munie A, Atkinson JR, Segal BM. Encephalitogenic and Regulatory CD8 T Cells in Multiple Sclerosis and Its Animal Models. THE JOURNAL OF IMMUNOLOGY 2021; 206:3-10. [PMID: 33443060 DOI: 10.4049/jimmunol.2000797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS), a neuroinflammatory disease that affects millions worldwide, is widely thought to be autoimmune in etiology. Historically, research into MS pathogenesis has focused on autoreactive CD4 T cells because of their critical role in the animal model, experimental autoimmune encephalomyelitis, and the association between MS susceptibility and single-nucleotide polymorphisms in the MHC class II region. However, recent studies have revealed prominent clonal expansions of CD8 T cells within the CNS during MS. In this paper, we review the literature on CD8 T cells in MS, with an emphasis on their potential effector and regulatory properties. We discuss the impact of disease modifying therapies, currently prescribed to reduce MS relapse rates, on CD8 T cell frequency and function. A deeper understanding of the role of CD8 T cells in MS may lead to the development of more effective and selective immunomodulatory drugs for particular subsets of patients.
Collapse
Affiliation(s)
- Taryn E Mockus
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Ashley Munie
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210; .,Neuroscience Research Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
35
|
Shahsavani N, Kataria H, Karimi-Abdolrezaee S. Mechanisms and repair strategies for white matter degeneration in CNS injury and diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166117. [PMID: 33667627 DOI: 10.1016/j.bbadis.2021.166117] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
White matter degeneration is an important pathophysiological event of the central nervous system that is collectively characterized by demyelination, oligodendrocyte loss, axonal degeneration and parenchymal changes that can result in sensory, motor, autonomic and cognitive impairments. White matter degeneration can occur due to a variety of causes including trauma, neurotoxic exposure, insufficient blood flow, neuroinflammation, and developmental and inherited neuropathies. Regardless of the etiology, the degeneration processes share similar pathologic features. In recent years, a plethora of cellular and molecular mechanisms have been identified for axon and oligodendrocyte degeneration including oxidative damage, calcium overload, neuroinflammatory events, activation of proteases, depletion of adenosine triphosphate and energy supply. Extensive efforts have been also made to develop neuroprotective and neuroregenerative approaches for white matter repair. However, less progress has been achieved in this area mainly due to the complexity and multifactorial nature of the degeneration processes. Here, we will provide a timely review on the current understanding of the cellular and molecular mechanisms of white matter degeneration and will also discuss recent pharmacological and cellular therapeutic approaches for white matter protection as well as axonal regeneration, oligodendrogenesis and remyelination.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
36
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
37
|
Xu Z, Lin CC, Ho S, Vlad G, Suciu-Foca N. Suppression of Experimental Autoimmune Encephalomyelitis by ILT3.Fc. THE JOURNAL OF IMMUNOLOGY 2020; 206:554-565. [PMID: 33361206 DOI: 10.4049/jimmunol.2000265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the CNS that is characterized by demyelination, axonal loss, gliosis, and inflammation. The murine model of MS is the experimental autoimmune encephalopathy (EAE) induced by immunization of mice with myelin oligodendrocyte glycoprotein (MOG)35-55 Ig-like transcript 3 (ILT3) is an inhibitory cell surface receptor expressed by tolerogenic human dendritic cells. In this study, we show that the recombinant human ILT3.Fc protein binds to murine immune cells and inhibits the release of proinflammatory cytokines that cause the neuroinflammatory process that result in paralysis. Administration of ILT3.Fc prevents the rapid evolution of the disease in C57BL/6 mice and is associated with a profound reduction of proliferation of MOG35-55-specific Th1 and Th17 cells. Inhibition of IFN-γ and IL-17A in mice treated with ILT3.Fc is associated with delayed time of onset of the disease and its evolution to a peak clinical score. Neuropathological analysis shows a reduction in inflammatory infiltrates and demyelinated areas in the brains and spinal cords of treated mice. These results indicate that inhibition of Th1 and Th17 development provides effective suppression of EAE and suggests the feasibility of a clinical approach based on the use of ILT3.Fc for treatment of MS. Furthermore, our results open the way to further studies on the effect of the human ILT3.Fc protein in murine experimental models of autoimmunity and cancer.
Collapse
Affiliation(s)
- Zheng Xu
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - Chun-Chieh Lin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Sophey Ho
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - George Vlad
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| | - Nicole Suciu-Foca
- Division of Immunogenetics and Cellular Immunology, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; and
| |
Collapse
|
38
|
Yu Z, Ling Z, Lu L, Zhao J, Chen X, Xu P, Zou X. Regulatory Roles of Bone in Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:610581. [PMID: 33408628 PMCID: PMC7779400 DOI: 10.3389/fnagi.2020.610581] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoporosis and neurodegenerative diseases are two kinds of common disorders of the elderly, which often co-occur. Previous studies have shown the skeletal and central nervous systems are closely related to pathophysiology. As the main structural scaffold of the body, the bone is also a reservoir for stem cells, a primary lymphoid organ, and an important endocrine organ. It can interact with the brain through various bone-derived cells, mostly the mesenchymal and hematopoietic stem cells (HSCs). The bone marrow is also a place for generating immune cells, which could greatly influence brain functions. Finally, the proteins secreted by bones (osteokines) also play important roles in the growth and function of the brain. This article reviews the latest research studying the impact of bone-derived cells, bone-controlled immune system, and bone-secreted proteins on the brain, and evaluates how these factors are implicated in the progress of neurodegenerative diseases and their potential use in the diagnosis and treatment of these diseases.
Collapse
Affiliation(s)
- Zhengran Yu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Lu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Zhao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
39
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
40
|
Lees JR. CD8+ T cells: The past and future of immune regulation. Cell Immunol 2020; 357:104212. [PMID: 32979764 DOI: 10.1016/j.cellimm.2020.104212] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 02/05/2023]
Abstract
Regulation of the adaptive immune response is critical for health. Regulatory activity can be found in multiple components of the immune system, however, the focus on particular components of the immune regulatory network has left many aspects of this critical immune component understudied. Here we review the evidence for activities of CD8+ T cells in immune homeostasis and regulation of autoimmune reactivity. The heterogeneous nature of identified CD8+ cell types are examined, and common phenotypes associated with functional activities are defined. The varying types of antigen signal crucial for CD8+ T cell regulatory activity are identified and the implications of these activation pathways for control of adaptive responses is considered. Finally, the promising capacity for transgenic antigen receptor directed cytotoxicity as a mechanism for modulation of autoimmunity is detailed.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
41
|
Wagner CA, Roqué PJ, Mileur TR, Liggitt D, Goverman JM. Myelin-specific CD8+ T cells exacerbate brain inflammation in CNS autoimmunity. J Clin Invest 2020; 130:203-213. [PMID: 31573979 DOI: 10.1172/jci132531] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the CNS. Although CD4+ T cells are implicated in MS pathogenesis and have been the main focus of MS research using the animal model experimental autoimmune encephalomyelitis (EAE), substantial evidence from patients with MS points to a role for CD8+ T cells in disease pathogenesis. We previously showed that an MHC class I-restricted epitope of myelin basic protein (MBP) is presented in the CNS during CD4+ T cell-initiated EAE. Here, we investigated whether naive MBP-specific CD8+ T cells recruited to the CNS during CD4+ T cell-initiated EAE engaged in determinant spreading and influenced disease. We found that the MBP-specific CD8+ T cells exacerbated brain but not spinal cord inflammation. We show that a higher frequency of monocytes and monocyte-derived cells presented the MHC class I-restricted MBP ligand in the brain compared with the spinal cord. Infiltration of MBP-specific CD8+ T cells enhanced ROS production in the brain only in these cell types and only when the MBP-specific CD8+ T cells expressed Fas ligand (FasL). These results suggest that myelin-specific CD8+ T cells may contribute to disease pathogenesis via a FasL-dependent mechanism that preferentially promotes lesion formation in the brain.
Collapse
Affiliation(s)
| | | | | | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
42
|
Benhammadi M, Mathé J, Dumont-Lagacé M, Kobayashi KS, Gaboury L, Brochu S, Perreault C. IFN-λ Enhances Constitutive Expression of MHC Class I Molecules on Thymic Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:1268-1280. [PMID: 32690660 DOI: 10.4049/jimmunol.2000225] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
Abstract
Regulation of MHC class I (MHC I) expression has been studied almost exclusively in hematolymphoid cells. We report that thymic epithelial cells (TECs), particularly the medullary TECs, constitutively express up to 100-fold more cell surface MHC I proteins than epithelial cells (ECs) from the skin, colon, and lung. Differential abundance of cell surface MHC I in primary ECs is regulated via transcription of MHC I and of genes implicated in the generation of MHC I-binding peptides. Superior MHC I expression in TECs is unaffected by deletion of Ifnar1 or Ifngr1, but is lessened by deletion of Aire, Ifnlr1, Stat1, or Nlrc5, and is driven mainly by type III IFN produced by medullary TECs. Ifnlr1 -/- mice show impaired negative selection of CD8 thymocytes and, at 9 mo of age, present autoimmune manifestations. Our study shows unanticipated variation in MHC I expression by ECs from various sites and provides compelling evidence that superior expression of MHC I in TECs is crucial for proper thymocyte education.
Collapse
Affiliation(s)
- Mohamed Benhammadi
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Justine Mathé
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Maude Dumont-Lagacé
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Koichi S Kobayashi
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, TX 77843.,Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan; and
| | - Louis Gaboury
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Sylvie Brochu
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; .,Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; .,Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
43
|
Abstract
Multiple sclerosis (MS) is an aggravating autoimmune disease that cripples young patients slowly with physical, sensory and cognitive deficits. The break of self-tolerance to neuronal antigens is the key to the pathogenesis of MS, with autoreactive T cells causing demyelination that subsequently leads to inflammation-mediated neurodegenerative events in the central nervous system. The exact etiology of MS remains elusive; however, the interplay of genetic and environmental factors contributes to disease development and progression. Given that genetic variation only accounts for a fraction of risk for MS, extrinsic risk factors including smoking, infection and lack of vitamin D or sunshine, which cause changes in gene expression, contribute to disease development through epigenetic regulation. To date, there is a growing body of scientific evidence to support the important roles of epigenetic processes in MS. In this chapter, the three main layers of epigenetic regulatory mechanisms, namely DNA methylation, histone modification and microRNA-mediated gene regulation, will be discussed, with a particular focus on the role of epigenetics on dysregulated immune responses and neurodegenerative events in MS. Also, the potential for epigenetic modifiers as biomarkers and therapeutics for MS will be reviewed.
Collapse
Affiliation(s)
- Vera Sau-Fong Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Queen Mary Hospital, Hong Kong SAR, China.
| |
Collapse
|
44
|
Chakravarty D, Saadi F, Kundu S, Bose A, Khan R, Dine K, Kenyon LC, Shindler KS, Das Sarma J. CD4 Deficiency Causes Poliomyelitis and Axonal Blebbing in Murine Coronavirus-Induced Neuroinflammation. J Virol 2020; 94:e00548-20. [PMID: 32404525 PMCID: PMC7343199 DOI: 10.1128/jvi.00548-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Mouse hepatitis virus (MHV) is a murine betacoronavirus (m-CoV) that causes a wide range of diseases in mice and rats, including hepatitis, enteritis, respiratory diseases, and encephalomyelitis in the central nervous system (CNS). MHV infection in mice provides an efficient cause-effect experimental model to understand the mechanisms of direct virus-induced neural-cell damage leading to demyelination and axonal loss, which are pathological features of multiple sclerosis (MS), the most common disabling neurological disease in young adults. Infiltration of T lymphocytes, activation of microglia, and their interplay are the primary pathophysiological events leading to disruption of the myelin sheath in MS. However, there is emerging evidence supporting gray matter involvement and degeneration in MS. The investigation of T cell function in the pathogenesis of deep gray matter damage is necessary. Here, we employed RSA59 (an isogenic recombinant strain of MHV-A59)-induced experimental neuroinflammation model to compare the disease in CD4-/- mice with that in CD4+/+ mice at days 5, 10, 15, and 30 postinfection (p.i.). Viral titer estimation, nucleocapsid gene amplification, and viral antinucleocapsid staining confirmed enhanced replication of the virions in the absence of functional CD4+ T cells in the brain. Histopathological analyses showed elevated susceptibility of CD4-/- mice to axonal degeneration in the CNS, with augmented progression of acute poliomyelitis and dorsal root ganglionic inflammation rarely observed in CD4+/+ mice. Depletion of CD4+ T cells showed unique pathological bulbar vacuolation in the brain parenchyma of infected mice with persistent CD11b+ microglia/macrophages in the inflamed regions on day 30 p.i. In summary, the current study suggests that CD4+ T cells are critical for controlling acute-stage poliomyelitis (gray matter inflammation), chronic axonal degeneration, and inflammatory demyelination due to loss of protective antiviral host immunity.IMPORTANCE The current trend in CNS disease biology is to attempt to understand the neural-cell-immune interaction to investigate the underlying mechanism of neuroinflammation, rather than focusing on peripheral immune activation. Most studies in MS are targeted toward understanding the involvement of CNS white matter. However, the importance of gray matter damage has become critical in understanding the long-term progressive neurological disorder. Our study highlights the importance of CD4+ T cells in safeguarding neurons against axonal blebbing and poliomyelitis from murine betacoronavirus-induced neuroinflammation. Current knowledge of the mechanisms that lead to gray matter damage in MS is limited, because the most widely used animal model, experimental autoimmune encephalomyelitis (EAE), does not present this aspect of the disease. Our results, therefore, add to the existing limited knowledge in the field. We also show that the microglia, though important for the initiation of neuroinflammation, cannot establish a protective host immune response without the help of CD4+ T cells.
Collapse
Affiliation(s)
- Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Abhishek Bose
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Reas Khan
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Kimberly Dine
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Lawrence C Kenyon
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kenneth S Shindler
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
- Department of Neurology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
45
|
Nichols JM, Kummari E, Sherman J, Yang EJ, Dhital S, Gilfeather C, Yray G, Morgan T, Kaplan BLF. CBD Suppression of EAE Is Correlated with Early Inhibition of Splenic IFN-γ + CD8+ T Cells and Modest Inhibition of Neuroinflammation. J Neuroimmune Pharmacol 2020; 16:346-362. [PMID: 32440886 DOI: 10.1007/s11481-020-09917-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/03/2020] [Indexed: 01/08/2023]
Abstract
In this study cannabidiol (CBD) was administered orally to determine its effects and mechanisms in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). We hypothesized that 75 mg/kg of oral CBD given for 5 days after initiation of disease would reduce EAE severity through suppression of either the early peripheral immune or late neuroimmune response. EAE was induced in C57BL/6 mice at two different magnitudes, and peripheral inflammatory and neuroinflammatory responses were measured at days 3, 10, and 18. Th1, Th17, Tc1, Tc17, Tregs, and myeloid derived suppressor cells (MDSC) were identified from the lymph nodes and spleens of each mouse to determine if CBD altered the suppressor cell or inflammatory cell populations in secondary lymphoid tissues. Additionally, neuroinflammation was identified in brain and spinal cord tissues using various immunohistochemical techniques and flow cytometry. Early treatment of EAE with oral CBD reduced clinical disease at the day 18 timepoint which correlated with a significant decrease in the percentage of MOG35-55 specific IFN-γ producing CD8+ T cells in the spleen at day 10. Analysis of both T cell infiltration and lesion size within the spinal cord also showed a moderate reduction in neuroinflammation within the central nervous system (CNS). These results provide evidence that oral CBD suppressed the peripheral immune response that precedes neuroinflammation; however, analysis of the neuroinflammatory endpoints also suggest that the modest reduction in neuroinflammation was only partially responsible for CBD's neuroprotective capability. Graphical Abstract CBD was administered orally for the first 5 days following initiation of EAE. CBD attenuated clinical disease, and we found that CBD suppressed IFN-γ producing CD8+ T cells in the spleen at day 10. There was also modest suppression of neuroinflammation. Together these data demonstrate that early, oral administration of CBD protected mice from disease, but the modest effects on neuroinflammation suggest other mechanisms participate in CBD's neuroprotective effect in EAE.
Collapse
Affiliation(s)
- James M Nichols
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Evangel Kummari
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Jessica Sherman
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Eun-Ju Yang
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Saphala Dhital
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Christa Gilfeather
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Gabriella Yray
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Timothy Morgan
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Barbara L F Kaplan
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA.
| |
Collapse
|
46
|
Choi BY, Jeong JH, Eom JW, Koh JY, Kim YH, Suh SW. A Novel Zinc Chelator, 1H10, Ameliorates Experimental Autoimmune Encephalomyelitis by Modulating Zinc Toxicity and AMPK Activation. Int J Mol Sci 2020; 21:ijms21093375. [PMID: 32397660 PMCID: PMC7247014 DOI: 10.3390/ijms21093375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 11/24/2022] Open
Abstract
Previous studies in our lab revealed that chemical zinc chelation or zinc transporter 3 (ZnT3) gene deletion suppresses the clinical features and neuropathological changes associated with experimental autoimmune encephalomyelitis (EAE). In addition, although protective functions are well documented for AMP-activated protein kinase (AMPK), paradoxically, disease-promoting effects have also been demonstrated for this enzyme. Recent studies have demonstrated that AMPK contributes to zinc-induced neurotoxicity and that 1H10, an inhibitor of AMPK, reduces zinc-induced neuronal death and protects against oxidative stress, excitotoxicity, and apoptosis. Here, we sought to evaluate the therapeutic efficacy of 1H10 against myelin oligodendrocyte glycoprotein 35-55-induced EAE. 1H10 (5 μg/kg) was intraperitoneally injected once per day for the entire experimental course. Histological evaluation was performed three weeks after the initial immunization. We found that 1H10 profoundly reduced the severity of the induced EAE and that there was a remarkable suppression of demyelination, microglial activation, and immune cell infiltration. 1H10 also remarkably inhibited EAE-associated blood-brain barrier (BBB) disruption, MMP-9 activation, and aberrant synaptic zinc patch formation. Furthermore, the present study showed that long-term treatment with 1H10 also reduced the clinical course of EAE. Therefore, the present study suggests that zinc chelation and AMPK inhibition with 1H10 may have great therapeutic potential for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Korea; (B.Y.C.); (J.H.J.)
| | - Jeong Hyun Jeong
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Korea; (B.Y.C.); (J.H.J.)
| | - Jae-Won Eom
- Department of Molecular Biology, Sejong University, Seoul 05006, Korea; (J.-W.E.); (Y.-H.K.)
| | - Jae-Young Koh
- Department of Neurology, University of Ulsan College of Medicine, Seoul 138-736, Korea;
| | - Yang-Hee Kim
- Department of Molecular Biology, Sejong University, Seoul 05006, Korea; (J.-W.E.); (Y.-H.K.)
| | - Sang Won Suh
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Korea; (B.Y.C.); (J.H.J.)
- Correspondence: ; Tel.: +82-10-8573-6364
| |
Collapse
|
47
|
Abdelsamed HA, Zebley CC, Nguyen H, Rutishauser RL, Fan Y, Ghoneim HE, Crawford JC, Alfei F, Alli S, Ribeiro SP, Castellaw AH, McGargill MA, Jin H, Boi SK, Speake C, Serti E, Turka LA, Busch ME, Stone M, Deeks SG, Sekaly RP, Zehn D, James EA, Nepom GT, Youngblood B. Beta cell-specific CD8 + T cells maintain stem cell memory-associated epigenetic programs during type 1 diabetes. Nat Immunol 2020; 21:578-587. [PMID: 32231298 PMCID: PMC7183435 DOI: 10.1038/s41590-020-0633-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/08/2020] [Indexed: 12/22/2022]
Abstract
The pool of beta cell-specific CD8+ T-cells in type 1 diabetes (T1D) sustains an autoreactive potential despite having access to a constant source of antigen. To investigate the long-lived nature of these cells, we established a DNA methylation-based T cell “multipotency index” and found that beta cell-specific CD8+ T-cells retained a stem-like epigenetic multipotency score. Single cell ATAC-seq analysis confirmed the co-existence of naive and effector-associated epigenetic programs in individual beta cell-specific CD8+ T-cells. Assessment of beta cell-specific CD8+ T-cell anatomical distribution and the establishment of stem-associated epigenetic programs revealed that self-reactive CD8+ T-cells isolated from murine lymphoid tissue retained developmentally plastic phenotypic and epigenetic profiles relative to the same cells isolated from the pancreas. Collectively, these data provide new insight into the longevity of beta cell-specific CD8+ T cell responses, and document the utility of this novel methylation-based multipotency index for investigating human and mouse CD8+ T-cell differentiation.
Collapse
Affiliation(s)
- Hossam A Abdelsamed
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Caitlin C Zebley
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hai Nguyen
- Translational Research Program, Benaroya Research Institute, Seattle, WA, USA
| | - Rachel L Rutishauser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hazem E Ghoneim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Francesca Alfei
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Shanta Alli
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Ashley H Castellaw
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shannon K Boi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cate Speake
- Diabetes Research Program, Benaroya Research Institute, Seattle, WA, USA
| | | | - Laurence A Turka
- Immune Tolerance Network, Bethesda, MD, USA.,Center for Translational Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Eddie A James
- Translational Research Program, Benaroya Research Institute, Seattle, WA, USA
| | - Gerald T Nepom
- Translational Research Program, Benaroya Research Institute, Seattle, WA, USA.,Immune Tolerance Network, Bethesda, MD, USA
| | - Ben Youngblood
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA. .,Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
48
|
Benkhoucha M, Senoner I, Lalive PH. c-Met is expressed by highly autoreactive encephalitogenic CD8+ cells. J Neuroinflammation 2020; 17:68. [PMID: 32075650 PMCID: PMC7031922 DOI: 10.1186/s12974-019-1676-0] [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: 09/26/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022] Open
Abstract
Background CD8+ T lymphocytes are critical mediators of neuroinflammatory diseases. Understanding the mechanisms that govern the function of this T cell population is crucial to better understanding central nervous system autoimmune disease pathology. We recently identified a novel population of highly cytotoxic c-Met-expressing CD8+ T lymphocytes and found that hepatocyte growth factor (HGF) limits effective murine cytotoxic T cell responses in cancer models. Here, we examined the role of c-Met-expressing CD8+ T cells by using a MOG35–55 T cell-mediated EAE model. Methods Mice were subcutaneously immunized with myelin oligodendrocyte glycoprotein peptide (MOG)35–55 in complete Freund’s adjuvant (CFA). Peripheral and CNS inflammation was evaluated at peak disease and chronic phase, and c-Met expression by CD8 was evaluated by flow cytometry and immunofluorescence. Molecular, cellular, and killing function analysis were performed by real-time PCR, ELISA, flow cytometry, and killing assay. Results In the present study, we observed that a fraction of murine effector CD8+ T cells expressed c-Met receptor (c-Met+CD8+) in an experimental autoimmune encephalitis (EAE) model. Phenotypic and functional analysis of c-Met+CD8+ T cells revealed that they recognize the encephalitogenic epitope myelin oligodendrocyte glycoprotein37–50. We demonstrated that this T cell population produces higher levels of interferon-γ and granzyme B ex vivo and that HGF directly restrains the cytolytic function of c-Met+CD8+ T cells in cell-mediated cytotoxicity reactions Conclusions Altogether, our findings suggest that the HGF/c-Met pathway could be exploited to modulate CD8+ T cell-mediated neuroinflammation.
Collapse
Affiliation(s)
- Mahdia Benkhoucha
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Isis Senoner
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrice H Lalive
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
49
|
Khaw YM, Cunningham C, Tierney A, Sivaguru M, Inoue M. Neutrophil-selective deletion of Cxcr2 protects against CNS neurodegeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2020; 17:49. [PMID: 32019585 PMCID: PMC7001284 DOI: 10.1186/s12974-020-1730-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/30/2020] [Indexed: 11/10/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic debilitating immune-mediated disease of the central nervous system (CNS) driven by demyelination and gray matter neurodegeneration. We previously reported an experimental autoimmune encephalomyelitis (EAE) MS mouse model with elevated serum CXCL1 that developed severe and prolonged neuron damage. Our findings suggested that CXCR2 signaling may be important in neuronal damage, thus implicating neutrophils, which express CXCR2 in abundance, as a potential cell type involved. The goals of this study were to determine if CXCR2 signaling in neutrophils mediate neuronal damage and to identify potential mechanisms of damage. Methods EAE was induced in wild-type control and neutrophil-specific Cxcr2 knockout (Cxcr2 cKO) mice by repeated high-dose injections of heat-killed Mycobacterium tuberculosis and MOG35–55 peptide. Mice were examined daily for motor deficit. Serum CXCL1 level was determined at different time points throughout disease development. Neuronal morphology in Golgi-Cox stained lumbar spinal cord ventral horn was assessed using recently developed confocal reflection super-resolution technique. Immune cells from CNS and lymphoid organs were quantified by flow cytometry. CNS-derived neutrophils were co-cultured with neuronal crest cells and neuronal cell death was measured. Neutrophils isolated from lymphoid organs were examined for expression of reactive oxygen species (ROS) and ROS-related genes. Thioglycolate-activated neutrophils were isolated, treated with recombinant CXCL1, and measured for ROS production. Results Cxcr2 cKO mice had less severe disease symptoms at peak and late phase when compared to control mice with similar levels of CNS-infiltrating neutrophils and other immune cells despite high levels of circulating CXCL1. Additionally, Cxcr2 cKO mice had significantly reduced CNS neuronal damage in the ventral horn of the spinal cord. Neutrophils isolated from control EAE mice induced vast neuronal cell death in vitro when compared with neutrophils isolated from Cxcr2 cKO EAE mice. Neutrophils isolated from control EAE mice, but not Cxcr2 cKO mice, exhibited elevated ROS generation, in addition to heightened Ncf1 and Il1b transcription. Furthermore, recombinant CXCL1 was sufficient to significantly increase neutrophils ROS production. Conclusions CXCR2 signal in neutrophils is critical in triggering CNS neuronal damage via ROS generation, which leads to prolonged EAE disease. These findings emphasize that CXCR2 signaling in neutrophils may be a viable target for therapeutic intervention against CNS neuronal damage.
Collapse
Affiliation(s)
- Yee Ming Khaw
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Claire Cunningham
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,The School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Abigail Tierney
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,The School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Mayandi Sivaguru
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Makoto Inoue
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
50
|
Fernández-Castañeda A, Chappell MS, Rosen DA, Seki SM, Beiter RM, Johanson DM, Liskey D, Farber E, Onengut-Gumuscu S, Overall CC, Dupree JL, Gaultier A. The active contribution of OPCs to neuroinflammation is mediated by LRP1. Acta Neuropathol 2020; 139:365-382. [PMID: 31552482 DOI: 10.1007/s00401-019-02073-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) account for about 5% of total brain and spinal cord cells, giving rise to myelinating oligodendrocytes that provide electrical insulation to neurons of the CNS. OPCs have also recently been shown to regulate inflammatory responses and glial scar formation, suggesting functions that extend beyond myelination. Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifaceted phagocytic receptor that is highly expressed in several CNS cell types, including OPCs. Here, we have generated an oligodendroglia-specific knockout of LRP1, which presents with normal myelin development, but is associated with better outcomes in two animal models of demyelination (EAE and cuprizone). At a mechanistic level, LRP1 did not directly affect OPC differentiation into mature oligodendrocytes. Instead, animals lacking LRP1 in OPCs in the demyelinating CNS were characterized by a robust dampening of inflammation. In particular, LRP1-deficient OPCs presented with impaired antigen cross-presentation machinery, suggesting a failure to propagate the inflammatory response and thus promoting faster myelin repair and neuroprotection. Our study places OPCs as major regulators of neuroinflammation in an LRP1-dependent fashion.
Collapse
Affiliation(s)
- Anthony Fernández-Castañeda
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Megan S Chappell
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Dorian A Rosen
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Graduate Program in Pharmacological Sciences, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Scott M Seki
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Rebecca M Beiter
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - David M Johanson
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Delaney Liskey
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Emily Farber
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Christopher C Overall
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Alban Gaultier
- Department of Neuroscience, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|