1
|
Chen JL, Wang B, Lu Y, Antoun E, Bird O, Drennan PG, Yin Z, Liu G, Yao X, Pidoux M, Bates A, Jayathilaka D, Wang J, Angus B, Beer S, Espinosa A, Baillie JK, Semple MG, Rostron T, Waugh C, Sopp P, Knight JC, Fullerton JN, Coles M, Smith GL, Mentzer AJ, Peng Y, Dong T. T cell memory response to MPXV infection exhibits greater effector function and migratory potential compared to MVA-BN vaccination. Nat Commun 2025; 16:4362. [PMID: 40348752 PMCID: PMC12065855 DOI: 10.1038/s41467-025-59370-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
In 2022, a global mpox outbreak occurred, and remains a concern today. The T cell memory response to MPXV (monkeypox virus) infection has not been fully investigated. In this study, we evaluate this response in convalescent and MVA-BN (Modified Vaccinia Ankara - Bavarian Nordic) vaccinated individuals using VACV-infected cells. Strong CD8+ and CD4+ T cell responses are observed, and T cell responses are biased towards viral early expressed proteins. We identify seven immunodominant HLA-A*02:01 restricted MPXV-specific epitopes and focus our detailed phenotypic and scRNAseq analysis on the immunodominant HLA-A*02:01-G5R18-26-specific CD8+ T cell response. While tetramer+CD8+ T cells share similar differentiation and activation phenotypes, T cells from convalescent individuals show greater cytotoxicity, migratory potential to site of infection and TCR clonal expansion. Our data suggest that effective functional profiles of MPXV-specific memory T cells induced by Mpox infection may have an implication on the long-term protective responses to future infection.
Collapse
Affiliation(s)
- Ji-Li Chen
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Beibei Wang
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Yongxu Lu
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Elie Antoun
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Olivia Bird
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Philip G Drennan
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Medicine, University of Oxford, Oxford, UK
| | - Zixi Yin
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Guihai Liu
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Xuan Yao
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Maya Pidoux
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Adam Bates
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Deshni Jayathilaka
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Junyuan Wang
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Brian Angus
- NDM Centre for Global Health Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sally Beer
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alexis Espinosa
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - J Kenneth Baillie
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
- Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
- Intensive Care Unit, Royal Infirmary Edinburgh, Edinburgh, UK
| | - Malcolm G Semple
- NIHR Health Protection Research Unit, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Timothy Rostron
- Sequencing Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Craig Waugh
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Paul Sopp
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Julian C Knight
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James N Fullerton
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Medicine, University of Oxford, Oxford, UK
| | - Mark Coles
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Medicine, University of Oxford, Oxford, UK
| | - Geoffrey L Smith
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Alexander J Mentzer
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yanchun Peng
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Tao Dong
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK.
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Bracken SJ, Poe JC, Sarantopoulos S. What's atypical about human B cells after allogeneic stem cell transplantation? J Leukoc Biol 2025; 117:qiaf048. [PMID: 40273381 PMCID: PMC12089796 DOI: 10.1093/jleuko/qiaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025] Open
Abstract
Atypical B cells or age-associated B cells represent an alternative lineage of memory B cells. Emerging evidence suggests that context influences the apparent functional heterogeneity of age-associated B cells. While data support a protective role for age-associated B cells in the setting of infection, multiple other studies suggest that these cells play a pathogenic role in the setting of autoimmunity. After treatment with allogeneic hematopoietic stem cell transplantation, the memory B-cell compartment is altered in patients who develop an autoimmune-like syndrome called chronic graft-versus-host disease. Patients with chronic graft-versus-host disease have significantly increased proportions of CD11c+ age-associated B cells within the peripheral compartment that develop under constant exposure to host alloantigens and persist under conditions when B-cell tolerance is not achieved. Herein, we review what is currently known about the molecular alterations in the heterogeneous memory B-cell compartment of hematopoietic stem cell transplantation patients, especially patients with chronic graft-versus-host disease who have developed autoimmune manifestations. In this mini-review, we summarize intrinsic factors in age-associated B cells found in autoimmune states that likely influence their extrafollicular localization, differentiation potential into autoantibody-secreting cells, and function. We highlight lessons from B-cell studies in chronic graft-versus-host disease to provide unique insights into the molecular underpinnings of the diverse functions of age-associated B cells.
Collapse
Affiliation(s)
- Sonali J Bracken
- Division of Rheumatology and Immunology, Duke University School of Medicine, 40 Duke Medicine Circle, Durham, NC 27710, United States
| | - Jonathan C Poe
- Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, 2400 Pratt Street, Durham, NC 27705, United States
| | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, 2400 Pratt Street, Durham, NC 27705, United States
- Department of Integrative Immunobiology, Duke University School of Medicine, 207 Research Drive Suite 156, Durham, NC 27706, United States
- Duke Cancer Institute, Duke University School of Medicine, DUMC Box 3917, Durham, NC 27710, United States
| |
Collapse
|
3
|
Gao L, Zhang Y, Hu Z, Chen S, Wang Q, Zeng Y, Yin H, Zhao J, Zhan Y, Gao C, Xin Y, Chen B, van der Veen S, Zhao M, Fang D, Lu Q. Microbiota-Derived Inosine Suppresses Systemic Autoimmunity via Restriction of B Cell Differentiation and Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409837. [PMID: 40289872 PMCID: PMC12120789 DOI: 10.1002/advs.202409837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/27/2025] [Indexed: 04/30/2025]
Abstract
The role of gut microbiota dysbiosis in systemic lupus erythematosus (SLE) pathogenesis remains elusive. Here, it is shown that fecal microbiota transplantation (FMT) from healthy mice to lupus mice ameliorates lupus-like symptoms. Microbiota reconstitution effectively reduces systemic class switch recombination (CSR) and elevates immunoglobulin heavy chain (IGH) naïve isotype. Microbiota profiling reveals an enrichment of Lactobacillus johnsonii post-FMT, with a significant correlation to purine metabolites. Importantly, the L. johnsonii-derived inosine, an intermediate metabolite in purine metabolism, effectively alleviates lupus pathogenesis in mice. Inosine inhibits B cell differentiation and reduces renal B cell infiltration to protect mice from lupus. At the molecular level, inosine reprograms B cells through the extracellular signal-regulated kinase (ERK)-hypoxia-inducible factor-1alpha (HIF-1α) signaling pathway. Therefore, this study highlights the discovery of a novel microbial metabolite modulating autoimmunity and suggests its potential for innovative microbiome-based therapeutic approaches.
Collapse
Affiliation(s)
- Lingyu Gao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yuhan Zhang
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Zhi Hu
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Shengwen Chen
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Qiaolin Wang
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yong Zeng
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| | - Huiqi Yin
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Junpeng Zhao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yijing Zhan
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Changxing Gao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Yue Xin
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
| | - Bing Chen
- Clinical LaboratoryThe Second Hospital of Anhui Medical UniversityHefei230601China
| | - Stijn van der Veen
- Department of Microbiologyand Department of DermatologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310058China
| | - Ming Zhao
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| | - Deyu Fang
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIL60611USA
| | - Qianjin Lu
- Hospital for Skin DiseasesInstitute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjing210042China
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjing210042China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIsNanjing210042China
- Hunan Key Laboratory of Medical EpigenomicsThe Second Xiangya HospitalCentral South UniversityChangsha410013China
| |
Collapse
|
4
|
Garcia-Coste JJ, Villafaña-Rauda S, Aguayo-Cerón KA, Vargas-De-León C, Romero-Nava R. KLF14 and SREBF-1 Binding Site Associations with Orphan Receptor Promoters in Metabolic Syndrome. Int J Mol Sci 2025; 26:2849. [PMID: 40243421 PMCID: PMC11988724 DOI: 10.3390/ijms26072849] [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: 12/16/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025] Open
Abstract
This study investigated the relationship between the transcription factors (TFs) KLF14 and SREBF-1 and orphan receptors (ORs) in the context of metabolic syndrome (MetS). A detailed bioinformatics analysis identified a significant association between the presence of binding sites (BS) for these TFs in the promoters of ORs genes and the total number of BS in the distal region. The results suggest that KLF14 and SREBF-1 can regulate the expression of some of these genes and, in turn, can modulate the development of MetS. Although a stronger association was observed with KLF14, both factors showed a significant contribution. Additionally, the sequence similarity of KLF14 also contributed to the quantity of BS in the gene's distal region (DR). The statistical models used, such as Poisson and negative binomial regression, confirmed these associations and allowed for the appropriate adjustment of overdispersion present in the data. However, no significant differences in receptor groups (orphan G Protein-Coupled Rereptors (oGPCRs) and G Protein-Coupled Receptors associated with MetS (GPCRs-MetS)) regarding their relationship with TFs were found. In conclusion, this study provides strong evidence of the importance of KLF14 and SREBF-1 in regulating orphan receptors genes and their participation in the development of metabolic syndrome.
Collapse
Affiliation(s)
- Julio Jesús Garcia-Coste
- Laboratorio de Investigación en Genética de Enfermedades Metabólicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico; (J.J.G.-C.); (K.A.A.-C.)
- Laboratorio de Modelación Bioestadística para la Salud, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico
| | - Santiago Villafaña-Rauda
- Laboratorio de Terapia Génica Experimental, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico;
| | - Karla Aidee Aguayo-Cerón
- Laboratorio de Investigación en Genética de Enfermedades Metabólicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico; (J.J.G.-C.); (K.A.A.-C.)
| | - Cruz Vargas-De-León
- Laboratorio de Modelación Bioestadística para la Salud, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico
- División de Investigación, Hospital Juárez de México, Ciudad de México 07760, Mexico
| | - Rodrigo Romero-Nava
- Laboratorio de Investigación en Genética de Enfermedades Metabólicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico; (J.J.G.-C.); (K.A.A.-C.)
| |
Collapse
|
5
|
Meloun A, León B. Beyond CCR7: dendritic cell migration in type 2 inflammation. Front Immunol 2025; 16:1558228. [PMID: 40093008 PMCID: PMC11906670 DOI: 10.3389/fimmu.2025.1558228] [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: 01/09/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Conventional dendritic cells (cDCs) are crucial antigen-presenting cells that initiate and regulate T cell responses, thereby shaping immunity against pathogens, innocuous antigens, tumors, and self-antigens. The migration of cDCs from peripheral tissues to draining lymph nodes (dLNs) is essential for their function in immune surveillance. This migration allows cDCs to convey the conditions of peripheral tissues to antigen-specific T cells in the dLNs, facilitating effective immune responses. Migration is primarily mediated by chemokine receptor CCR7, which is upregulated in response to homeostatic and inflammatory cues, guiding cDCs to dLNs. However, during type 2 immune responses, such as those triggered by parasites or allergens, a paradox arises-cDCs exhibit robust migration to dLNs despite low CCR7 expression. This review discusses how type 2 inflammation relies on additional signaling pathways, including those induced by membrane-derived bioactive lipid mediators like eicosanoids, sphingolipids, and oxysterols, which cooperate with CCR7 to enhance cDC migration and T helper 2 (Th2) differentiation. We explore the potential regulatory mechanisms of cDC migration in type 2 immunity, offering insights into the differential control of cDC trafficking in diverse immune contexts and its impact on immune responses.
Collapse
Affiliation(s)
- Audrey Meloun
- Innate Cells and Th2 Immunity Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Beatriz León
- Innate Cells and Th2 Immunity Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
6
|
Caratis F, Karaszewski B, Klejbor I, Furihata T, Rutkowska A. Differential expression and modulation of EBI2 and 7α,25-OHC synthesizing (CH25H, CYP7B1) and degrading (HSD3B7) enzymes in mouse and human brain vascular cells. PLoS One 2025; 20:e0318822. [PMID: 39999050 PMCID: PMC11856462 DOI: 10.1371/journal.pone.0318822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The endogenous ligand for the EBI2 receptor, oxysterol 7α,25OHC, crucial for immune responses, is finely regulated by CH25H, CYP7B1 and HSD3B7 enzymes. Lymphoid stromal cells and follicular dendritic cells within T cell follicles maintain a gradient of 7α,25OHC, with stromal cells increasing and dendritic cells decreasing its concentration. This gradient is pivotal for proper B cell positioning in lymphoid tissue. In the animal model of multiple sclerosis, the experimental autoimmune encephalomyelitis, the levels of 7α,25OHC rapidly increase in the central nervous system driving the migration of EBI2 expressing immune cells through the blood-brain barrier (BBB). To explore if blood vessel cells in the brain express these enzymes, we examined normal mouse brain microvessels and studied changes in their expression during inflammation. Ebi2 was abundantly expressed in endothelial cells, pericytes/smooth muscle cells, and astrocytic endfeet. Ch25h, Cyp7b1, and Hsd3b7 were variably detected in each cell type, suggesting their active involvement in oxysterol 7α,25OHC synthesis and gradient maintenance under normal conditions. Significant species-specific differences emerged in EBI2 and the enzyme levels between mouse and human BBB-forming cells. Under acute inflammatory conditions, Ebi2 and synthesizing enzyme modulation occurred in the brain, with the magnitude and direction of change based on the enzyme. Lastly, in an in vitro astrocyte migration model, CYP7B1 inhibitor clotrimazole, as well as EBI2 antagonist, NIBR189, inhibited lipopolysaccharide-induced cell migration indicating the involvement of EBI2 and its ligand in brain cell migration under inflammatory conditions.
Collapse
Affiliation(s)
- Fionä Caratis
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
| | - Bartosz Karaszewski
- Department of Adult Neurology, Medical University of Gdansk & University Clinical Center, Gdansk, Poland
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
| | - Ilona Klejbor
- Department of Anatomy, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Tomomi Furihata
- Laboratory of Clinical Pharmacy and Experimental Therapeutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Aleksandra Rutkowska
- Department of Anatomy and Neurobiology, Medical University of Gdansk, Gdansk, Poland
- Brain Diseases Centre, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
7
|
Chen KY, De Giovanni M, Xu Y, An J, Kirthivasan N, Lu E, Jiang K, Brooks S, Ranucci S, Yang J, Kanameishi S, Kabashima K, Brulois K, Bscheider M, Butcher EC, Cyster JG. Inflammation switches the chemoattractant requirements for naive lymphocyte entry into lymph nodes. Cell 2025; 188:1019-1035.e22. [PMID: 39708807 PMCID: PMC11845304 DOI: 10.1016/j.cell.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/27/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Sustained lymphocyte migration from blood into lymph nodes (LNs) is important for immune responses. The CC-chemokine receptor-7 (CCR7) ligand CCL21 is required for LN entry but is downregulated during inflammation, and it has been unclear how recruitment is maintained. Here, we show that the oxysterol biosynthetic enzyme cholesterol-25-hydroxylase (Ch25h) is upregulated in LN high endothelial venules during viral infection. Lymphocytes become dependent on oxysterols, generated through a transcellular endothelial-fibroblast metabolic pathway, and the receptor EBI2 for inflamed LN entry. Additionally, Langerhans cells are an oxysterol source. Ch25h is also expressed in inflamed peripheral endothelium, and EBI2 mediates B cell recruitment in a tumor model. Finally, we demonstrate that LN CCL19 is critical in lymphocyte recruitment during inflammation. Thus, our work explains how naive precursor trafficking is sustained in responding LNs, identifies a role for oxysterols in cell recruitment into inflamed tissues, and establishes a logic for the CCR7 two-ligand system.
Collapse
Affiliation(s)
- Kevin Y Chen
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nikhita Kirthivasan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erick Lu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kan Jiang
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Serena Ranucci
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jiuling Yang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shuto Kanameishi
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kevin Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Michael Bscheider
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Palacios PA, Santibañez Á, Aguirre-Muñoz F, Gutiérrez-Vera C, Niño de Zepeda-Carrizo V, Góngora-Pimentel M, Müller M, Cáceres M, Kalergis AM, Carreño LJ. Can invariant Natural Killer T cells drive B cell fate? a look at the humoral response. Front Immunol 2025; 16:1505883. [PMID: 40040714 PMCID: PMC11876049 DOI: 10.3389/fimmu.2025.1505883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Invariant Natural Killer T (NKT) cells represent a unique subset of innate-like T cells that express both NK cell and T cell receptors. These cells are rapidly activated by glycolipid antigens presented via CD1d molecules on antigen-presenting cells (APCs), including B cells, dendritic cells (DCs), and macrophages, or through cytokine-dependent mechanisms. Their ability to produce a wide range of cytokines and express costimulatory molecules underscores their critical role in bridging innate and adaptive immunity. B cells, traditionally recognized for their role in antibody production, also act as potent APCs due to their high expression of CD1d, enabling direct interactions with iNKT cells. This interaction has significant implications for humoral immunity, influencing B cell activation, class-switch recombination (CSR), germinal center formation, and memory B cell differentiation, thus expanding the conventional paradigm of T cell-B cell interactions. While the influence of iNKT cells on B cell biology and humoral responses is well-supported, many aspects of their interaction remain unresolved. Key questions include the roles of different iNKT cell subsets, the diversity of APCs, the spatiotemporal dynamics of these interactions, especially during early activation, and the potential for distinct glycolipid ligands to modulate immune outcomes. Understanding these factors could provide valuable insights into how iNKT cells regulate B cell-mediated immunity and offer opportunities to harness these interactions in immunotherapeutic applications, such as vaccine development. In this review, we examine these unresolved aspects and propose a novel perspective on the regulatory potential of iNKT cells in humoral immunity, emphasizing their promise as a target for innovative vaccine strategies.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Álvaro Santibañez
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernanda Aguirre-Muñoz
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Niño de Zepeda-Carrizo
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Martín Góngora-Pimentel
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
9
|
Zhou Y, Hubscher CH. Biomarker expression level changes within rectal gut-associated lymphoid tissues in spinal cord-injured rats. Immunohorizons 2025; 9:vlaf002. [PMID: 40048710 PMCID: PMC11884801 DOI: 10.1093/immhor/vlaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurogenic bowel dysfunction (NBD) is common after spinal cord injury (SCI). Gut-associated lymphoid tissue (GALT), an organized structure within the mucosal immune system, is important for the maintenance of gut homeostasis and body health and serves as the first line barrier/defense against diet antigens, commensal microbiota, pathogens, and toxins in mucosal areas. The current study examined gene expression levels along six segments of anorectal tissue using real-time polymerase chain reaction (RT-PCR) in uninjured rats (28-day sham surgical controls) and at both 28- and 42-days post-T9 contusion injury. Consistent with our previous report of functional regional differences in the ano-rectum, we demonstrate the existence of GALTs located primarily within the segment at 3-4.5 cm from the rectal dentate line (termed rectal GALTs-rGALTs) in shams with upregulated gene expression levels of multiple biomarkers, including B cell and T cell-related genes, major histocompatibility complex (MHC) class II molecules, and germinal center (GC)-related genes, which was further confirmed by histologic examination. In the same rectal tissue segment following T9 SCI, inflammation-related genes were upregulated at 28 days post-injury (DPI) indicating that microbial infection and inflammation of rGALTs modified structure and function of rGALTs, while at 42 DPI rGALTs exhibited resolution of inflammation and impaired structure/function for extrafollicular B cell responses. Taken together, our data suggest that rGALTs exists in rat rectum for homeostasis of gut microbiota/barrier. SCI induces microbial infection and inflammation in rectal tissues containing rGALTs, which could contribute to development of SCI-related gut microbiome dysbiosis, NBD, and systemic diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| |
Collapse
|
10
|
Kim Y, Manara F, Grassmann S, Belcheva KT, Reyes K, Kim H, Downs-Canner S, Yewdell WT, Sun JC, Chaudhuri J. IL-21 shapes the B cell response in a context-dependent manner. Cell Rep 2025; 44:115190. [PMID: 39792552 PMCID: PMC11973891 DOI: 10.1016/j.celrep.2024.115190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
The T-cell-derived cytokine IL-21 is crucial for germinal center (GC) responses, but its precise role in B cell function has remained elusive. Using IL-21 receptor (Il21r) conditional knockout mice and ex vivo culture systems, we demonstrate that IL-21 has dual effects on B cells. While IL-21 induced apoptosis in a STAT3-dependent manner in naive B cells, it promoted the robust proliferation of pre-activated B cells, particularly IgG1+ B cells. In vivo, B-cell-specific Il21r deletion impaired IgG1 responses post-immunization and disrupted progression from pre-GC to GC states. Although Il21r deficiency did not affect the proportion of IgG1+ cells among GC B cells, it greatly diminished the proportion of IgG1+ cells among the plasmablast/plasma cell population. Collectively, our findings suggest that IL-21 serves as a critical regulator of B cell fates, influencing B cell apoptosis and proliferation in a context-dependent manner.
Collapse
Affiliation(s)
- Youngjun Kim
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.
| | - Francesca Manara
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kalina T Belcheva
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Kanelly Reyes
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hyunu Kim
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | | | - William T Yewdell
- Department of Immunology Discovery, Genentech Inc, South San Francisco, CA 94080, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | - Jayanta Chaudhuri
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY 10065, USA.
| |
Collapse
|
11
|
Zhuang H, Li F, Pei R, Jiang X, Chen D, Li S, Ye P, Yuan J, Lian J, Jin J, Lu Y. High Expression of GPR183 Predicts Poor Survival in Cytogenetically Normal Acute Myeloid Leukemia. Biochem Genet 2025:10.1007/s10528-025-11026-1. [PMID: 39820826 DOI: 10.1007/s10528-025-11026-1] [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: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Acute myeloid leukemia (AML) with a normal karyotype (CN-AML) constitutes approximately 50% of all AML cases, presenting significant prognostic variability, and highlighting the urgent need for the identification of novel molecular biomarkers. In this study, we systematically assessed GPR183 expression levels using qRT-PCR in our clinical follow-up study which included 283 CN-AML patients. Using Kaplan-Meier analysis, we found that patients with high GPR183 expression levels exhibited significantly worse overall survival (OS) (P = 0.046) and event-free survival (EFS) (P = 0.030) compared to those with low GPR183 expression. Comprehensive univariate and multivariate Cox regression analyses confirmed that GPR183 expression is a prognostic factor for OS and EFS (P < 0.05). To further validate these findings, we analyzed an independent cohort of 104 CN-AML patients from the GSE71014 dataset, corroborating our primary results, and indicating that high GPR183 expression is associated with poorer survival outcomes. Additionally, RNA-seq data from the GSE71014 dataset were analyzed by Gene Set Enrichment Analysis (GSEA). The results suggested that GPR183 may influence disease progression through the activation of the "TNFa Signaling Via NF-κB" pathway. Collectively, these findings suggested that GPR183 could serve as a valuable prognostic biomarker in CN-AML, offering insights into the underlying mechanisms of disease progression.
Collapse
Affiliation(s)
- Haihui Zhuang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Fenglin Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Xia Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Dong Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Peipei Ye
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jiaojiao Yuan
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jiangyin Lian
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
- Institute of Hematology, Ningbo University, Ningbo, China
| | - Jie Jin
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, PR China.
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China.
- Institute of Hematology, Ningbo University, Ningbo, China.
| |
Collapse
|
12
|
Liu Y, Qin J, Li X, Wu G. Oxysterols in tumor immune microenvironment (TIME). J Steroid Biochem Mol Biol 2025; 245:106634. [PMID: 39551164 DOI: 10.1016/j.jsbmb.2024.106634] [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: 07/29/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Oxysterols are compounds generated through oxidative reactions involving cholesterol and other steroid molecules. They play a crucial role in the tumor immune microenvironment by interacting with molecules such as the cell membrane receptor EBI2 and nuclear receptors like LXR and PXR. This interaction regulates immune cell signaling pathways, affecting proliferation, apoptosis, migration, and invasion in tumor-related processes. Activating these receptors alters the function and behavior of immune cells-such as macrophages, T cells, and dendritic cells-within the tumor microenvironment, thus promoting or inhibiting tumor development. Certain oxidized steroids can increase both the number and activation of infiltrating T cells, synergizing with anti-PD-1 to enhance anti-tumor efficacy. An in-depth study of the biological mechanisms of oxidized sterols will not only enhance our understanding of the complexity of the tumor immune microenvironment but may also reveal new therapeutic targets, providing innovative strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuanxin Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Jie Qin
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
13
|
Konieczna-Wolska K, Caratis F, Opiełka M, Biernacki K, Urbanowicz K, Klimaszewska J, Pobiarzyn P, Krajewski O, Demkowicz S, Smoleński RT, Karaszewski B, Seuwen K, Rutkowska A. Accelerated remyelination and immune modulation by the EBI2 agonist 7α,25-dihydroxycholesterol analogue in the cuprizone model. Biomed Pharmacother 2024; 181:117653. [PMID: 39489122 DOI: 10.1016/j.biopha.2024.117653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024] Open
Abstract
Research indicates a role for EBI2 receptor in remyelination, demonstrating that its deficiency or antagonism inhibits this process. However, activation of EBI2 with its endogenous ligand, oxysterol 7α,25-dihydroxycholesterol (7α,25OHC), does not enhance remyelination beyond the levels observed in spontaneously remyelinating tissue. We hypothesized that the short half-life of the natural ligand might explain this lack of beneficial effects and tested a synthetic analogue, CF3-7α,25OHC, in the cuprizone model. The data showed that extending the bioavailability of 7α,25OHC is sufficient to accelerate remyelination in vivo. Moreover, the analogue, in contrast to the endogenous ligand, upregulated brain expression of Ebi2 and the synthesis of 15 lipids in the mouse corpus callosum. Mechanistically, the increased concentration of oxysterol likely disrupted its gradient in demyelinated areas of the brain, leading to the dispersion of infiltrating EBI2-expressing immune cells rather than their accumulation in demyelinated regions. Remarkably, the analogue CF3-7α,25OHC markedly decreased the lymphocyte and monocyte counts mimicking the key mechanism of action of some of the most effective disease-modifying therapies for multiple sclerosis. Furthermore, the Cd4+ transcripts in the cerebellum and CD4+ cell number in the corpus callosum were reduced compared to vehicle-treated mice. These findings suggest a mechanism by which EBI2/7α,25OHC signalling modulates the immune response and accelerates remyelination in vivo.
Collapse
Affiliation(s)
- Klaudia Konieczna-Wolska
- Brain Diseases Centre, Medical University of Gdańsk, Gdańsk, Poland; Tri-City Central Animal Laboratory Research and Service Center, Medical University of Gdańsk, Gdańsk, Poland
| | - Fionä Caratis
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland; Department of Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Mikołaj Opiełka
- Brain Diseases Centre, Medical University of Gdańsk, Gdańsk, Poland; Department of Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | - Joanna Klimaszewska
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Pobiarzyn
- Brain Diseases Centre, Medical University of Gdańsk, Gdańsk, Poland
| | - Oliwier Krajewski
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | - Bartosz Karaszewski
- Brain Diseases Centre, Medical University of Gdańsk, Gdańsk, Poland; Department of Adult Neurology, Medical University of Gdańsk and University Clinical Center, Gdańsk, Poland
| | - Klaus Seuwen
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Aleksandra Rutkowska
- Brain Diseases Centre, Medical University of Gdańsk, Gdańsk, Poland; Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
14
|
von Voss L, Arora T, Assis J, Kuentzel KB, Arfelt KN, Nøhr MK, Grevengoed TJ, Arumugam M, Mandrup-Poulsen T, Rosenkilde MM. Sexual Dimorphism in the Immunometabolic Role of Gpr183 in Mice. J Endocr Soc 2024; 8:bvae188. [PMID: 39545055 PMCID: PMC11561910 DOI: 10.1210/jendso/bvae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Indexed: 11/17/2024] Open
Abstract
Context Excessive eating and intake of a Western diet negatively affect the intestinal immune system, resulting in compromised glucose homeostasis and lower gut bacterial diversity. The G protein-coupled receptor GPR183 regulates immune cell migration and intestinal immune response and has been associated with tuberculosis, type 1 diabetes, and inflammatory bowel diseases. Objective We hypothesized that with these implications, GPR183 has an important immunometabolic role and investigated this using a global Gpr183 knockout mouse model. Methods Wild-type (WT) and Gpr183-deficient (Gpr183-/-) mice were fed a high-fat, high-sucrose diet (HFSD) for 15 weeks. We investigated changes in weight, body composition, fecal immunoglobulin A (IgA) levels, fecal microbiome, and glucose tolerance before and after the diet. Macrophage infiltration into visceral fat was determined by flow cytometry, and hepatic gene expression was measured. Results A sexual dimorphism was discovered, whereby female Gpr183-/- mice showed adverse metabolic outcomes compared to WT counterparts with inferior glucose tolerance, lower fecal IgA levels, and increased macrophage infiltration in visceral fat. In contrast, male Gpr183-/- mice had significantly lower fasting blood glucose after diet than male WT mice. Liver gene expression showed reduced inflammation and macrophage markers in Gpr183-/- livers, regardless of sex, while the pancreatic islet area did not differ between the groups. No conclusive differences were found after microbiome sequencing. Conclusion Gpr183 maintains metabolic homeostasis in female but not in male mice independent of diet. If confirmed in humans, future therapy targeting GPR183 should consider this sexual dimorphism.
Collapse
Affiliation(s)
- Liv von Voss
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Juliana Assis
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Immunotechnology, Lund University, SE 223 63 Lund, Sweden
| | - Katharina B Kuentzel
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Kristine N Arfelt
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Mark K Nøhr
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Trisha J Grevengoed
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Mette M Rosenkilde
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| |
Collapse
|
15
|
Back P, Yu M, Modaresahmadi S, Hajimirzaei S, Zhang Q, Islam MR, Schwendeman AA, La-Beck NM. Immune Implications of Cholesterol-Containing Lipid Nanoparticles. ACS NANO 2024; 18:28480-28501. [PMID: 39388645 PMCID: PMC11505898 DOI: 10.1021/acsnano.4c06369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
The majority of clinically approved nanoparticle-mediated therapeutics are lipid nanoparticles (LNPs), and most of these LNPs are liposomes containing cholesterol. LNP formulations significantly alter the drug pharmacokinetics (PK) due to the propensity of nanoparticles for uptake by macrophages. In addition to readily engulfing LNPs, the high expression of cholesterol hydroxylases and reactive oxygen species (ROS) in macrophages suggests that they will readily produce oxysterols from LNP-associated cholesterol. Oxysterols are a heterogeneous group of cholesterol oxidation products that have potent immune modulatory effects. Oxysterols are implicated in the pathogenesis of atherosclerosis and certain malignancies; they have also been found in commercial liposome preparations. Yet, the in vivo metabolic fate of LNP-associated cholesterol remains unclear. We review herein the mechanisms of cellular uptake, trafficking, metabolism, and immune modulation of endogenous nanometer-sized cholesterol particles (i.e., lipoproteins) that are also relevant for cholesterol-containing nanoparticles. We believe that it would be imperative to better understand the in vivo metabolic fate of LNP-associated cholesterol and the immune implications for LNP-therapeutics. We highlight critical knowledge gaps that we believe need to be addressed in order to develop safer and more efficacious lipid nanoparticle delivery systems.
Collapse
Affiliation(s)
- Patricia
Ines Back
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Minzhi Yu
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Shadan Modaresahmadi
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Sahelosadat Hajimirzaei
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Qisheng Zhang
- Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Md Rakibul Islam
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Anna A. Schwendeman
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
- Biointerfaces
Institute, University of Michigan, North
Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Ninh M. La-Beck
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
- Department
of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| |
Collapse
|
16
|
Morgan DM, Zhang YJ, Kim JH, Murillo M, Singh S, Loschko J, Surendran N, Sekulovic O, Feng E, Shi S, Irvine DJ, Patil SU, Kanevsky I, Chorro L, Christopher Love J. Full-length single-cell BCR sequencing paired with RNA sequencing reveals convergent responses to pneumococcal vaccination. Commun Biol 2024; 7:1208. [PMID: 39341987 PMCID: PMC11438910 DOI: 10.1038/s42003-024-06823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) can resolve transcriptional features from individual cells, but scRNA-seq techniques capable of resolving the variable regions of B cell receptors (BCRs) remain limited, especially from widely-used 3'-barcoded libraries. Here, we report a method that can recover paired, full-length variable region sequences of BCRs from 3'-barcoded scRNA-seq libraries. We first verify this method (B3E-seq) can produce accurate, full-length BCR sequences. We then apply this method to profile B cell responses elicited against the capsular polysaccharide of Streptococcus pneumoniae serotype 3 (ST3) by glycoconjugate vaccines in five infant rhesus macaques. We identify BCR features associated with specificity for the ST3 antigen which are present in multiple vaccinated monkeys, indicating a convergent response to vaccination. These results demonstrate the utility of our method to resolve key features of the B cell repertoire and profile antigen-specific responses elicited by vaccination.
Collapse
Affiliation(s)
- Duncan M Morgan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Yiming J Zhang
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Jin-Hwan Kim
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - MaryAnn Murillo
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Suddham Singh
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Jakob Loschko
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Deerfield Management, New York, NY, USA
| | - Naveen Surendran
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ognjen Sekulovic
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ellie Feng
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Shuting Shi
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Materials Science and Engineering, MIT, Cambridge, MA, USA
| | - Sarita U Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isis Kanevsky
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Laurent Chorro
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Regeneron, Tarrytown, NY, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- Department of Chemical Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
17
|
Bai J, Kato A, Hulse KE, Wechsler JB, Gujar V, Poposki JA, Harmon R, Iwasaki N, Wang BF, Huang JH, Stevens WW, Conley DB, Welch KC, Kern RC, Peters AT, Eisenbarth SC, Schleimer RP, Tan BK. Increased autoreactivity and maturity of EBI2+ antibody-secreting cells from nasal polyps. JCI Insight 2024; 9:e177729. [PMID: 39253973 PMCID: PMC11385095 DOI: 10.1172/jci.insight.177729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/18/2024] [Indexed: 09/11/2024] Open
Abstract
Elevated numbers of antibody-secreting cells (ASCs) and anti-double-stranded DNA (anti-dsDNA) antibodies are found in nasal polyp (NP) tissue. The presence of anti-dsDNA IgG in tissue prospectively predicts recurrent NP but the characteristics of the source ASCs are unknown. Here, we investigated whether NP B cells expressing the extrafollicular marker EBI2 have increased propensity for autoantibody production and evaluated the molecular characteristics of NP ASCs. NPs showed increased frequencies of anti-dsDNA IgG and total IgG ASCs compared with tonsils, with more pronounced differences among EBI2+ cells. In NPs, EBI2+ cells were frequently double negative (IgD-CD27-) and ASCs. Single-cell RNA-Seq analysis of tonsils and NPs revealed substantial differences in B lineage composition, including differences in percentages of ASCs, germinal centers, proliferative cells, and non-ASCs. NPs exhibited higher expression of specific isotypes (IGHE, IGHA1, IGHA2, and IGHG4) and mature plasma genes, including SDC1 and XBP1, than tonsils. Gene Ontology biological processes indicated upregulated NF-κB and downregulated apoptosis pathways in NP ASCs. Together, these data indicate that NP EBI2+ ASCs secret increased total and anti-dsDNA IgG compared with those from tonsils and had molecular features of mature plasma cell differentiation.
Collapse
Affiliation(s)
| | - Atsushi Kato
- Department of Otolaryngology
- Division of Allergy and Immunology, Department of Medicine, and
| | | | - Joshua B. Wechsler
- Departments of Pediatrics and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Vikram Gujar
- Department of Anatomy and Cell Biology, Oklahoma State University, Tulsa, Oklahoma, USA
| | | | | | | | - Bao-Feng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Wuhan, China
| | | | - Whitney W. Stevens
- Department of Otolaryngology
- Division of Allergy and Immunology, Department of Medicine, and
| | | | | | | | - Anju T. Peters
- Department of Otolaryngology
- Division of Allergy and Immunology, Department of Medicine, and
| | | | - Robert P. Schleimer
- Department of Otolaryngology
- Division of Allergy and Immunology, Department of Medicine, and
| | - Bruce K. Tan
- Department of Otolaryngology
- Division of Allergy and Immunology, Department of Medicine, and
| |
Collapse
|
18
|
Cinti I, Vezyrgianni K, Denton AE. Unravelling the contribution of lymph node fibroblasts to vaccine responses. Adv Immunol 2024; 164:1-37. [PMID: 39523027 DOI: 10.1016/bs.ai.2024.07.001] [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: 11/16/2024]
Abstract
Vaccination is one of the most effective medical interventions, saving millions of lives and reducing the morbidity of infections across the lifespan, from infancy to older age. The generation of plasma cells and memory B cells that produce high affinity class switched antibodies is central to this protection, and these cells are the ultimate output of the germinal centre response. Optimal germinal centre responses require different immune cells to interact with one another in the right place and at the right time and this delicate cellular ballet is coordinated by a network of interconnected stromal cells. In this review we will discuss the various types of lymphoid stromal cells and how they coordinate immune cell homeostasis, the induction and maintenance of the germinal centre response, and how this is disorganised in older bodies.
Collapse
Affiliation(s)
- Isabella Cinti
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Kassandra Vezyrgianni
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Alice E Denton
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom.
| |
Collapse
|
19
|
Guigues A, Gimenez S, Mettling C, Maurel D, Doumazane E, Prézeau L, François V, Corbeau P. The EBI2 receptor is coexpressed with CCR5 in CD4 + T cells and boosts HIV-1 R5 replication. AIDS 2024; 38:1449-1459. [PMID: 38770825 DOI: 10.1097/qad.0000000000003931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
OBJECTIVE CCR5, a G protein-coupled receptor (GPCR), is used by most HIV strains as a coreceptor. In this study, we looked for other GPCR able to modify HIV-1 infection. DESIGN We analyzed the effects of one GPCR coexpressed with CCR5, EBI2, on HIV-1 replicative cycle. METHODS We identified GPCR expressed in primary CD4 + CCR5 + T cells by multi-RT-qPCR. We studied GPCR dimerization by FRET technology. Cell lines expressing EBI2 were established by transduction with HIV vectors. HIV-1 entry was quantified with virions harboring β-lactamase fused to the viral protein vpr, early and late HIV-1 transcriptions by qPCR, NFkB nuclear activation by immunofluorescence and transfection, and viral production by measuring p24 concentration in culture supernatant by ELISA. RESULTS We showed that EBI2 is naturally expressed in primary CD4 + CCR5 + T cells, and that CCR5 and EBI2 heterodimerize. We observed that this coexpression reduced viral entry by 50%. The amount of HIV reverse transcripts was similar in cells expressing or not EBI2. Finally, the presence of EBI2 induced the translocation of NFkB and activated HIV-1 genome expression. Globally, the result was a drastic HIV-1 R5, but not X4, overproduction in EBI2 -transduced cells. CONCLUSION EBI2 expression in CD4 + CCR5 + cells boosts HIV-1 R5 productive infection. As the natural ligand for EBI2 is present in blood and lymphoid tissues, the constant EBI2 activation might increase HIV replication in CD4 + T cells. It might be of interest to test the effect of EBI2 antagonists on the residual viral production persisting in patients aviremic under treatment.
Collapse
Affiliation(s)
- Adeline Guigues
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002
| | - Sandrine Gimenez
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002
| | - Clément Mettling
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002
| | - Damien Maurel
- ARPEGE Pharmacology Screening Interactome Platform Facility
| | - Etienne Doumazane
- Institut de Génomique Fonctionnelle, CNRS-UMR5203, INSERM-U661, Universités Montpellier 1 & 2
- Paris Brain Institute (ICM), Sorbonne Université, INSERM U1127, CNRS UMR7225, Paris, France
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, CNRS-UMR5203, INSERM-U661, Universités Montpellier 1 & 2
| | - Vincent François
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002
| | - Pierre Corbeau
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002
- Université de Montpellier
- Centre Hospitalier Universitaire Carémeau, UF d'Immunologie, Nîmes Cedex 9
| |
Collapse
|
20
|
Kim Y, Manara F, Grassmann S, Belcheva KT, Reyes K, Kim H, Downs-Canner S, Yewdell WT, Sun JC, Chaudhuri J. IL-21 Shapes the B Cell Response in a Context-Dependent Manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.600808. [PMID: 39026745 PMCID: PMC11257567 DOI: 10.1101/2024.07.13.600808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The cytokine interleukin-21 (IL-21) is a pivotal T cell-derived signal crucial for germinal center (GC) responses, but the precise mechanisms by which IL-21 influences B cell function remain elusive. Here, we investigated the B cell-intrinsic role of IL-21 signaling by employing a novel IL-21 receptor ( Il21r ) conditional knock-out mouse model and ex vivo culture systems and uncovered a surprising duality of IL-21 signaling in B cells. While IL-21 stimulation of naïve B cells led to Bim-dependent apoptosis, it promoted robust proliferation of pre-activated B cells, particularly class-switched IgG1 + B cells ex vivo . Consistent with this, B cell-specific deletion of Il21r led to a severe defect in IgG1 responses in vivo following immunization. Intriguingly, Il21r -deleted B cells are significantly impaired in their ability to transition from a pre-GC to a GC state following immunization. Although Il21r -deficiency did not affect the proportion of IgG1 + B cells among GC B cells, it greatly diminished the proportion of IgG1 + B cells among the plasmablast/plasma cell population. Collectively, our data suggest that IL-21 serves as a critical regulator of B cell fates, influencing B cell apoptosis and proliferation in a context-dependent manner.
Collapse
|
21
|
Hu L, An K, Zhang Y, Bai C. Exploring the Activation Mechanism of the GPR183 Receptor. J Phys Chem B 2024; 128:6071-6081. [PMID: 38877985 DOI: 10.1021/acs.jpcb.4c02812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The G protein-coupled receptors (GPCRs) play a pivotal role in numerous biological processes as crucial cell membrane receptors. However, the dynamic mechanisms underlying the activation of GPR183, a specific GPCR, remain largely elusive. To address this, we employed computational simulation techniques to elucidate the activation process and key events associated with GPR183, including conformational changes from inactive to active state, binding interactions with the Gi protein complex, and GDP release. Our findings demonstrate that the association between GPR183 and the Gi protein involves the formation of receptor-specific conformations, the gradual proximity of the Gi protein to the binding pocket, and fine adjustments of the protein conformation, ultimately leading to a stable GPR183-Gi complex characterized by a high energy barrier. The presence of Gi protein partially promotes GPR183 activation, which is consistent with the observation of GPCR constitutive activity test experiments, thus illustrating the reliability of our calculations. Moreover, our study suggests the existence of a stable partially activated state preceding complete activation, providing novel avenues for future investigations. In addition, the relevance of GPR183 for various diseases, such as colitis, the response of eosinophils to Mycobacterium tuberculosis infection, antiviral properties, and pulmonary inflammation, has been emphasized, underscoring its therapeutic potential. Consequently, understanding the activation process of GPR183 through molecular dynamic simulations offers valuable kinetic insights that can aid in the development of targeted therapies.
Collapse
Affiliation(s)
- Linfeng Hu
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China
- Warshel Institute for Computational Biology, Shenzhen, Guangdong 518172, PR China
| | - Ke An
- Chenzhu (MoMeD) Biotechnology Co., Ltd, Hangzhou, Zhejiang 310005, PR China
| | - Yue Zhang
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China
- Warshel Institute for Computational Biology, Shenzhen, Guangdong 518172, PR China
| | - Chen Bai
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China
- Warshel Institute for Computational Biology, Shenzhen, Guangdong 518172, PR China
- Chenzhu (MoMeD) Biotechnology Co., Ltd, Hangzhou, Zhejiang 310005, PR China
| |
Collapse
|
22
|
Zeng R, Fang M, Shen A, Chai X, Zhao Y, Liu M, Zhu L, Rui W, Feng B, Hong L, Ding C, Song Z, Lu W, Zhang A. Discovery of a Highly Potent Oxysterol Receptor GPR183 Antagonist Bearing the Benzo[ d]thiazole Structural Motif for the Treatment of Inflammatory Bowel Disease (IBD). J Med Chem 2024; 67:3520-3541. [PMID: 38417036 DOI: 10.1021/acs.jmedchem.3c01905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Accumulating evidence has demonstrated a critical pathological role of oxysterol receptor GPR183 in various inflammatory and autoimmune diseases, including inflammatory bowel disease (IBD). However, the currently reported GPR183 antagonists are very limited and not qualified for in vivo studies due to their inferior druglike properties. Herein, we conducted a structural elaboration focusing on improving its PK and safety profile based on a reference antagonist NIBR189. Of note, compound 33, bearing an aminobenzothiazole motif, exhibited reduced hERG inhibition, improved PK properties, and robust antagonistic activity (IC50 = 0.82 nM) with high selectivity against GPR183. Moreover, compound 33 displayed strong in vitro antimigration and anti-inflammatory activity in monocytes. Oral administration of compound 33 effectively improved the pathological symptoms of DSS-induced experimental colitis. All of these findings demonstrate that compound 33 is a novel and promising GPR183 antagonist suitable for further investigation to treat IBD.
Collapse
Affiliation(s)
- Ruoqing Zeng
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
| | - Meimiao Fang
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ancheng Shen
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Lingang Laboratory, Shanghai 200210, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
| | - Xiaolei Chai
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yumiao Zhao
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lingfeng Zhu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Weiwei Rui
- Department of General Surgery and Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Bo Feng
- Department of General Surgery and Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Liang Hong
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
| | - Chunyong Ding
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
| | - Zilan Song
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ao Zhang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Lingang Laboratory, Shanghai 200210, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai 200240, China
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
23
|
He Y, Vinuesa CG. Germinal center versus extrafollicular responses in systemic autoimmunity: Who turns the blade on self? Adv Immunol 2024; 162:109-133. [PMID: 38866437 PMCID: PMC7616122 DOI: 10.1016/bs.ai.2024.02.002] [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] [Indexed: 06/14/2024]
Abstract
Spontaneously formed germinal centers (GCs) have been reported in most mouse models of human autoimmune disease and autoimmune patients, and have long been considered a source of somatically-mutated and thus high affinity autoantibodies, but their role in autoimmunity is becoming increasingly controversial, particularly in the context of systemic autoimmune diseases like lupus. On the one hand, there is good evidence that some pathogenic lupus antibodies have acquired somatic mutations that increase affinity for self-antigens. On the other hand, recent studies that have genetically prevented GC formation, suggest that GCs are dispensable for systemic autoimmunity, pointing instead to pathogenic extrafollicular (EF) B-cell responses. Furthermore, several lines of evidence suggest germinal centers may in fact be somewhat protective in the context of autoimmunity. Here we review how some of the conflicting evidence arose, and current views on the role of GCs in autoimmunity, outlining mechanisms by which GC may eliminate self-reactivity. We also discuss recent advances in understanding extrafollicular B cell subsets that participate in autoimmunity.
Collapse
Affiliation(s)
- Yuke He
- China-Australia Centre for Personalised Immunology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Carola G Vinuesa
- China-Australia Centre for Personalised Immunology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China; Francis Crick Institute, London, United Kingdom.
| |
Collapse
|
24
|
Callegari I, Oechtering J, Schneider M, Perriot S, Mathias A, Voortman MM, Cagol A, Lanner U, Diebold M, Holdermann S, Kreiner V, Becher B, Granziera C, Junker A, Du Pasquier R, Khalil M, Kuhle J, Kappos L, Sanderson NSR, Derfuss T. Cell-binding IgM in CSF is distinctive of multiple sclerosis and targets the iron transporter SCARA5. Brain 2024; 147:839-848. [PMID: 38123517 PMCID: PMC10907079 DOI: 10.1093/brain/awad424] [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: 04/05/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 12/23/2023] Open
Abstract
Intrathecal IgM production in multiple sclerosis is associated with a worse disease course. To investigate pathogenic relevance of autoreactive IgM in multiple sclerosis, CSF from two independent cohorts, including multiple sclerosis patients and controls, were screened for antibody binding to induced pluripotent stem cell-derived neurons and astrocytes, and a panel of CNS-related cell lines. IgM binding to a primitive neuro-ectodermal tumour cell line discriminated 10% of multiple sclerosis donors from controls. Transcriptomes of single IgM producing CSF B cells from patients with cell-binding IgM were sequenced and used to produce recombinant monoclonal antibodies for characterization and antigen identification. We produced five cell-binding recombinant IgM antibodies, of which one, cloned from an HLA-DR + plasma-like B cell, mediated antigen-dependent complement activation. Immunoprecipitation and mass spectrometry, and biochemical and transcriptome analysis of the target cells identified the iron transport scavenger protein SCARA5 as the antigen target of this antibody. Intrathecal injection of a SCARA5 antibody led to an increased T cell infiltration in an experimental autoimmune encephalomyelitis (EAE) model. CSF IgM might contribute to CNS inflammation in multiple sclerosis by binding to cell surface antigens like SCARA5 and activating complement, or by facilitating immune cell migration into the brain.
Collapse
Affiliation(s)
- Ilaria Callegari
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Johanna Oechtering
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Mika Schneider
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Sylvain Perriot
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
| | - Amandine Mathias
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
| | | | - Alessandro Cagol
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel 4123, Switzerland
| | - Ulrike Lanner
- Proteomics Core Facility, Biozentrum, University of Basel, Basel 4056, Switzerland
| | - Martin Diebold
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg 79085, Germany
| | - Sebastian Holdermann
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Victor Kreiner
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Cristina Granziera
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel 4123, Switzerland
| | - Andreas Junker
- Department of Neuropathology, University Hospital Essen, Essen 45147, Germany
| | - Renaud Du Pasquier
- Department of Clinical Neurosciences, Laboratory of Neuroimmunology, Center of Research in Neurosciences, Lausanne 1011, Switzerland
- Department of Clinical Neurosciences, Service of Neurology, Lausanne 1011, Switzerland
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz 8010, Austria
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Ludwig Kappos
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Nicholas S R Sanderson
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| | - Tobias Derfuss
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel 4056, Switzerland
| |
Collapse
|
25
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
26
|
Lanzolla G, Comi S, Cosentino G, Pakdel F, Marinò M. Statins in Graves Orbitopathy: A New Therapeutic Tool. Ophthalmic Plast Reconstr Surg 2023; 39:S29-S39. [PMID: 38054983 DOI: 10.1097/iop.0000000000002525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
PURPOSE Graves orbitopathy (GO) is the most common extrathyroidal manifestation of Graves disease. Although its pathogenesis is not fully elucidated, GO is commonly considered an autoimmune disease due to loss of self-tolerance against autoantigens shared by thyroid epithelial cells and orbital fibroblasts. High-dose intravenous glucocorticoids (ivGCs) are the most used treatment for moderate-to-severe, active GO, but the addition of other immunomodulating treatments can improve the efficacy of ivGCs. Among the various risk factors that can affect the occurrence of GO, cholesterol may be worthy of interest. Since 2015 the role of cholesterol and cholesterol-lowering medications has been investigated. The purpose of this review is to discuss this topic, thereby offering new therapeutic opportunities for patients with GO. METHODS We searched PubMed for studies published between January 1, 1980 and June 1, 2023, using the search terms "Graves orbitopathy," "thyroid eye disease," "Graves ophthalmopathy," "thyroid ophthalmopathy," "thyroid-associated ophthalmopathy," "endocrine ophthalmopathy," "cholesterol," "lipids," "statins," "low-density lipoprotein," "atorvastatin," and "cholesterol-lowering drugs." Only English-language articles were included. RESULTS A correlation between low-density lipoprotein cholesterol and the risk of GO development has been reported. Furthermore, low-density lipoprotein cholesterol has been proposed as a risk factor that can affect the course of GO and the response to ivGCs. The protective role of cholesterol-lowering medications in preventing GO has been also investigated. Statin treatment was found to have potential benefits in reducing the risk of GO in patients with Graves disease. Given these findings, measurement of low-density lipoprotein cholesterol and treatment of hypercholesterolemia in patients with moderate-to-severe, active GO may be considered before starting ivGCs administration. Recently, a randomized clinical trial aimed at investigating the effects of statins in GO suggested that the addition of oral atorvastatin to ivGCs improves the overall outcome of moderate-to-severe, active GO in hypercholesterolemic patients given ivGCs. CONCLUSIONS Overall, statins seem to have a preventive and therapeutic role in moderate-to-severe active GO. Their efficacy can be related to cholesterol-lowering activity, pleiotropic actions, and interaction with methylprednisolone.
Collapse
Affiliation(s)
- Giulia Lanzolla
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Simone Comi
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Giada Cosentino
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Farzad Pakdel
- Department of Ophthalmic Plastic and Reconstructive Surgery, Farabi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Michele Marinò
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
27
|
Ding Z, Mulder J, Robinson MJ. The origins and longevity of IgE responses as indicated by serological and cellular studies in mice and humans. Allergy 2023; 78:3103-3117. [PMID: 37417548 PMCID: PMC10952832 DOI: 10.1111/all.15799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
The existence of long-lived IgE antibody-secreting cells (ASC) is contentious, with the maintenance of sensitization by the continuous differentiation of short-lived IgE+ ASC a possibility. Here, we review the epidemiological profile of IgE production, and give an overview of recent discoveries made on the mechanisms regulating IgE production from mouse models. Together, these data suggest that for most individuals, in most IgE-associated diseases, IgE+ ASC are largely short-lived cells. A subpopulation of IgE+ ASC in humans is likely to survive for tens of months, although due to autonomous IgE B cell receptor (BCR) signaling and antigen-driven IgE+ ASC apoptosis, in general IgE+ ASC probably do not persist for the decades that other ASC are inferred to do. We also report on recently identified memory B cell transcriptional subtypes that are the likely source of IgE in ongoing responses, highlighting the probable importance of IL-4Rα in their regulation. We suggest the field should look at dupilumab and other drugs that prohibit IgE+ ASC production as being effective treatments for IgE-mediated aspects of disease in most individuals.
Collapse
Affiliation(s)
- Zhoujie Ding
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | - Jesse Mulder
- Department of ImmunologyMonash UniversityMelbourneVictoriaAustralia
| | | |
Collapse
|
28
|
Kjær VMS, Stępniewski TM, Medel-Lacruz B, Reinmuth L, Ciba M, Rexen Ulven E, Bonomi M, Selent J, Rosenkilde MM. Ligand entry pathways control the chemical space recognized by GPR183. Chem Sci 2023; 14:10671-10683. [PMID: 37829039 PMCID: PMC10566501 DOI: 10.1039/d2sc05962b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 08/26/2023] [Indexed: 10/14/2023] Open
Abstract
The G protein-coupled receptor GPR183 is a chemotactic receptor with an important function in the immune system and association with a variety of diseases. It recognizes ligands with diverse physicochemical properties as both the endogenous oxysterol ligand 7α,25-OHC and synthetic molecules can activate the G protein pathway of the receptor. To better understand the ligand promiscuity of GPR183, we utilized both molecular dynamics simulations and cell-based validation experiments. Our work reveals that the receptor possesses two ligand entry channels: one lateral between transmembrane helices 4 and 5 facing the membrane, and one facing the extracellular environment. Using enhanced sampling, we provide a detailed structural model of 7α,25-OHC entry through the lateral membrane channel. Importantly, the first ligand recognition point at the receptor surface has been captured in diverse experimentally solved structures of different GPCRs. The proposed ligand binding pathway is supported by in vitro data employing GPR183 mutants with a sterically blocked lateral entrance, which display diminished binding and signaling. In addition, computer simulations and experimental validation confirm the existence of a polar water channel which might serve as an alternative entrance gate for less lipophilic ligands from the extracellular milieu. Our study reveals knowledge to understand GPR183 functionality and ligand recognition with implications for the development of drugs for this receptor. Beyond, our work provides insights into a general mechanism GPCRs may use to respond to chemically diverse ligands.
Collapse
Affiliation(s)
- Viktoria Madeline Skovgaard Kjær
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen Blegdamsvej 3B 2200 København N Denmark
| | - Tomasz Maciej Stępniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM) & Pompeu Fabra University (UPF) Dr Aiguader 88 E-8003 Barcelona Spain
- InterAx Biotech AG, PARK innovAARE 5234 Villigen Switzerland
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw 02-089 Warsaw Poland
| | - Brian Medel-Lacruz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM) & Pompeu Fabra University (UPF) Dr Aiguader 88 E-8003 Barcelona Spain
| | - Lisa Reinmuth
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen Blegdamsvej 3B 2200 København N Denmark
| | - Marija Ciba
- Department of Drug Design and Pharmacology, University of Copenhagen Jagtvej 160 2100 København Ø Denmark
| | - Elisabeth Rexen Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen Jagtvej 160 2100 København Ø Denmark
| | - Massimiliano Bonomi
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit 75015 Paris France
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM) & Pompeu Fabra University (UPF) Dr Aiguader 88 E-8003 Barcelona Spain
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen Blegdamsvej 3B 2200 København N Denmark
| |
Collapse
|
29
|
Shi Q, Zhan T, Bi X, Ye BC, Qi N. Cholesterol-autoxidation metabolites in host defense against infectious diseases. Eur J Immunol 2023; 53:e2350501. [PMID: 37369622 DOI: 10.1002/eji.202350501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
Cholesterol plays essential roles in biological processes, including cell membrane stability and myelin formation. Cholesterol can be metabolized to oxysterols by enzymatic or nonenzymatic ways. Nonenzymatic cholesterol metabolites, also called cholesterol-autoxidation metabolites, are formed dependent on the oxidation of reactive oxygen species (ROS) such as OH• or reactive nitrogen species, such as ONOO- . Cholesterol-autoxidation metabolites are abundantly produced in diseases such as inflammatory bowel disease and atherosclerosis, which are associated with oxidative stress. Recent studies have shown that cholesterol-autoxidation metabolites can further regulate the immune system. Here, we review the literature and summarize how cholesterol-autoxidation metabolites, such as 25-hydroxycholesterol (25-OHC), 7α/β-OHC, and 7-ketocholesterol, deal with the occurrence and development of infectious diseases through pattern recognition receptors, inflammasomes, ROS production, nuclear receptors, G-protein-coupled receptor 183, and lipid availability. In addition, we include the research regarding the roles of these metabolites in COVID-19 infection and discuss our viewpoints on the future research directions.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Tingzhu Zhan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Xiaobao Bi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Bang-Ce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Nan Qi
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Laboratory, Department of Basic Research, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Zhang F, Zhang B, Ding H, Li X, Wang X, Zhang X, Liu Q, Feng Q, Han M, Chen L, Qi L, Yang D, Li X, Zhu X, Zhao Q, Qiu J, Zhu Z, Tang H, Shen N, Wang H, Wei B. The Oxysterol Receptor EBI2 Links Innate and Adaptive Immunity to Limit IFN Response and Systemic Lupus Erythematosus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207108. [PMID: 37469011 PMCID: PMC10520634 DOI: 10.1002/advs.202207108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/19/2023] [Indexed: 07/21/2023]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease with abnormal activation of the immune system. Recent attention is increasing about how aberrant lipid and cholesterol metabolism is linked together with type I interferon (IFN-I) signaling in the regulation of the pathogenesis of SLE. Here, a metabonomic analysis is performed and increased plasma concentrations of oxysterols, especially 7α, 25-dihydroxycholesterol (7α, 25-OHC), are identified in SLE patients. The authors find that 7α, 25-OHC binding to its receptor Epstein-Barr virus-induced gene 2 (EBI2) in macrophages can suppress STAT activation and the production of IFN-β, chemokines, and cytokines. Importantly, monocytes/macrophages from SLE patients and mice show significantly reduced EBI2 expression, which can be triggered by IFN-γ produced in activated T cells. Previous findings suggest that EBI2 enhances immune cell migration. Opposite to this effect, the authors demonstrate that EBI2-deficient macrophages produce higher levels of chemokines and cytokines, which recruits and activates myeloid cells,T and B lymphocytes to exacerbate tetramethylpentadecane-induced SLE. Together, via sensing the oxysterol 7α, 25-OHC, EBI2 in macrophages can modulate innate and adaptive immune responses, which may be used as a potential diagnostic marker and therapeutic target for SLE.
Collapse
Affiliation(s)
- Fang Zhang
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
- Cancer CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Baokai Zhang
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Huihua Ding
- Shanghai Institute of RheumatologyRenji HospitalShanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200127China
| | - Xiangyue Li
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Xilin Wang
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Xiaomin Zhang
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesUniversity of Chinese Academy of ScienceWuhan430071China
| | - Qiaojie Liu
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesUniversity of Chinese Academy of ScienceWuhan430071China
| | - Qiuyun Feng
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Mingshun Han
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Longlong Chen
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhangjiang Fudan International Innovation CenterZhongshan HospitalFudan UniversityShanghai200032China
- Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular PhenomicsFudan UniversityShanghai200032China
| | - Linlin Qi
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Dan Yang
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesUniversity of Chinese Academy of ScienceWuhan430071China
| | - Xiaojing Li
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Xingguo Zhu
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Qi Zhao
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Jiaqian Qiu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai200032China
| | - Zhengjiang Zhu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghai200032China
| | - Huiru Tang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhangjiang Fudan International Innovation CenterZhongshan HospitalFudan UniversityShanghai200032China
- Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular PhenomicsFudan UniversityShanghai200032China
| | - Nan Shen
- Shanghai Institute of RheumatologyRenji HospitalShanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200127China
| | - Hongyan Wang
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
- School of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| | - Bin Wei
- Institute of GeriatricsAffiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong)School of MedicineShanghai UniversityNantong226011China
- Immune Cells and Human Diseases Lab, Shanghai Engineering Research Center of Organ RepairSchool of Life SciencesShanghai UniversityShanghai200444China
- Cancer CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesUniversity of Chinese Academy of ScienceWuhan430071China
- Department of Laboratory MedicineGene Diagnosis Research CenterFujian Key Laboratory of Laboratory MedicineThe First Affiliated HospitalFujian Medical UniversityFuzhou350000China
| |
Collapse
|
31
|
Matz H, Dooley H. 450 million years in the making: mapping the evolutionary foundations of germinal centers. Front Immunol 2023; 14:1245704. [PMID: 37638014 PMCID: PMC10450919 DOI: 10.3389/fimmu.2023.1245704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Germinal centers (GCs) are distinct microanatomical structures that form in the secondary lymphoid organs of endothermic vertebrates (i.e., mammals and some birds). Within GCs, B cells undergo a Darwinian selection process to identify clones which can respond to pathogen insult as well as affinity mature the B cell repertoire. The GC response ultimately generates memory B cells and bone marrow plasma cells which facilitate humoral immunological memory, the basis for successful vaccination programs. GCs have not been observed in the secondary lymphoid organs of ectothermic jawed vertebrates (i.e., fishes, reptiles, and amphibians). However, abundant research over the past decades has indicated these organisms can produce antigen specific B cell responses and some degree of affinity maturation. This review examines data demonstrating that the fundamentals of B cell selection may be more conserved across vertebrate phylogeny than previously anticipated. Further, research in both conventional mammalian model systems and comparative models raises the question of what evolutionary benefit GCs provide endotherms if they are seemingly unnecessary for generating the basic functional components of jawed vertebrate humoral adaptive immune responses.
Collapse
|
32
|
Matz HC, McIntire KM, Ellebedy AH. 'Persistent germinal center responses: slow-growing trees bear the best fruits'. Curr Opin Immunol 2023; 83:102332. [PMID: 37150126 PMCID: PMC10829534 DOI: 10.1016/j.coi.2023.102332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/09/2023]
Abstract
Germinal centers (GCs) are key microanatomical sites in lymphoid organs where responding B cells mature and undergo affinity-based selection. The duration of the GC reaction has long been assumed to be relatively brief, but recent studies in humans, nonhuman primates, and mice indicate that GCs can last for weeks to months after initial antigen exposure. This review examines recent studies investigating the factors that influence GC duration, including antigen persistence, T-follicular helper cells, and mode of immunization. Potential mechanisms for how persistent GCs influence the B-cell repertoire are considered. Overall, these studies provide a blueprint for how to design better vaccines that elicit persistent GC responses.
Collapse
Affiliation(s)
- Hanover C Matz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, USA.
| |
Collapse
|
33
|
Matz H, Taylor RS, Redmond AK, Hill TM, Ruiz Daniels R, Beltran M, Henderson NC, Macqueen DJ, Dooley H. Organized B cell sites in cartilaginous fishes reveal the evolutionary foundation of germinal centers. Cell Rep 2023; 42:112664. [PMID: 37342909 PMCID: PMC10529500 DOI: 10.1016/j.celrep.2023.112664] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 02/28/2023] [Accepted: 06/04/2023] [Indexed: 06/23/2023] Open
Abstract
The absence of germinal centers (GCs) in cartilaginous fishes lies at odds with data showing that nurse sharks can produce robust antigen-specific responses and affinity mature their B cell repertoires. To investigate this apparent incongruity, we performed RNA sequencing on single nuclei, allowing us to characterize the cell types present in the nurse shark spleen, and RNAscope to provide in situ cellular resolution of key marker gene expression following immunization with R-phycoerythrin (PE). We tracked PE to the splenic follicles where it co-localizes with CXCR5high centrocyte-like B cells and a population of putative T follicular helper (Tfh) cells, surrounded by a peripheral ring of Ki67+ AID+ CXCR4+ centroblast-like B cells. Further, we reveal selection of mutations in B cell clones dissected from these follicles. We propose that the B cell sites identified here represent the evolutionary foundation of GCs, dating back to the jawed vertebrate ancestor.
Collapse
Affiliation(s)
- Hanover Matz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, MD, USA
| | - Richard S Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Anthony K Redmond
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Thomas M Hill
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, MD, USA
| | - Rose Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Mariana Beltran
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Helen Dooley
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, MD, USA.
| |
Collapse
|
34
|
Lütge M, De Martin A, Gil-Cruz C, Perez-Shibayama C, Stanossek Y, Onder L, Cheng HW, Kurz L, Cadosch N, Soneson C, Robinson MD, Stoeckli SJ, Ludewig B, Pikor NB. Conserved stromal-immune cell circuits secure B cell homeostasis and function. Nat Immunol 2023; 24:1149-1160. [PMID: 37202489 PMCID: PMC10307622 DOI: 10.1038/s41590-023-01503-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 04/03/2023] [Indexed: 05/20/2023]
Abstract
B cell zone reticular cells (BRCs) form stable microenvironments that direct efficient humoral immunity with B cell priming and memory maintenance being orchestrated across lymphoid organs. However, a comprehensive understanding of systemic humoral immunity is hampered by the lack of knowledge of global BRC sustenance, function and major pathways controlling BRC-immune cell interactions. Here we dissected the BRC landscape and immune cell interactome in human and murine lymphoid organs. In addition to the major BRC subsets underpinning the follicle, including follicular dendritic cells, PI16+ RCs were present across organs and species. As well as BRC-produced niche factors, immune cell-driven BRC differentiation and activation programs governed the convergence of shared BRC subsets, overwriting tissue-specific gene signatures. Our data reveal that a canonical set of immune cell-provided cues enforce bidirectional signaling programs that sustain functional BRC niches across lymphoid organs and species, thereby securing efficient humoral immunity.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | | | - Yves Stanossek
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
- Department of Otorhinolaryngology Head and Neck Surgery, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Charlotte Soneson
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Sandro J Stoeckli
- Department of Otorhinolaryngology Head and Neck Surgery, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland.
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland.
| |
Collapse
|
35
|
Gonzales C, Liang Y, Fisher J, Card G, Sun J, Soong L. Alterations in germinal center formation and B cell activation during severe Orientia tsutsugamushi infection in mice. PLoS Negl Trop Dis 2023; 17:e0011090. [PMID: 37146079 PMCID: PMC10191367 DOI: 10.1371/journal.pntd.0011090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/17/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
Scrub typhus is a poorly studied but life-threatening disease caused by the intracellular bacterium Orientia tsutsugamushi (Ot). Cellular and humoral immunity in Ot-infected patients is not long-lasting, waning as early as one-year post-infection; however, its underlying mechanisms remain unclear. To date, no studies have examined germinal center (GC) or B cell responses in Ot-infected humans or experimental animals. This study was aimed at evaluating humoral immune responses at acute stages of severe Ot infection and possible mechanisms underlying B cell dysfunction. Following inoculation with Ot Karp, a clinically dominant strain known to cause lethal infection in C57BL/6 mice, we measured antigen-specific antibody titers, revealing IgG2c as the dominant isotype induced by infection. Splenic GC responses were evaluated by immunohistology, co-staining for B cells (B220), T cells (CD3), and GCs (GL-7). Organized GCs were evident at day 4 post-infection (D4), but they were nearly absent at D8, accompanied by scattered T cells throughout splenic tissues. Flow cytometry revealed comparable numbers of GC B cells and T follicular helper (Tfh) cells at D4 and D8, indicating that GC collapse was not due to excessive death of these cell subtypes at D8. B cell RNAseq analysis revealed significant differences in expression of genes associated with B cell adhesion and co-stimulation at D8 versus D4. The significant downregulation of S1PR2 (a GC-specific adhesion gene) was most evident at D8, correlating with disrupted GC formation. Signaling pathway analysis uncovered downregulation of 71% of B cell activation genes at D8, suggesting attenuation of B cell activation during severe infection. This is the first study showing the disruption of B/T cell microenvironment and dysregulation of B cell responses during Ot infection, which may help understand the transient immunity associated with scrub typhus.
Collapse
Affiliation(s)
- Casey Gonzales
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - James Fisher
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Galen Card
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
36
|
Kjær VMS, Daugvilaite V, Stepniewski TM, Madsen CM, Jørgensen AS, Bhuskute KR, Inoue A, Ulven T, Benned-Jensen T, Hjorth SA, Hjortø GM, Moo EV, Selent J, Rosenkilde MM. Migration mediated by the oxysterol receptor GPR183 depends on arrestin coupling but not receptor internalization. Sci Signal 2023; 16:eabl4283. [PMID: 37014928 DOI: 10.1126/scisignal.abl4283] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
The chemotactic G protein-coupled receptor GPR183 and its most potent endogenous oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) are important for immune cell positioning in secondary lymphoid tissues. This receptor-ligand pair is associated with various diseases, in some cases contributing favorably and in other cases adversely, making GPR183 an attractive target for therapeutic intervention. We investigated the mechanisms underlying GPR183 internalization and the role of internalization in the main biological function of the receptor, chemotaxis. We found that the C terminus of the receptor was important for ligand-induced internalization but less so for constitutive (ligand-independent) internalization. β-arrestin potentiated ligand-induced internalization but was not required for ligand-induced or constitutive internalization. Caveolin and dynamin were the main mediators of both constitutive and ligand-induced receptor internalization in a mechanism independent of G protein activation. Clathrin-mediated endocytosis also contributed to constitutive GPR183 internalization in a β-arrestin-independent manner, suggesting the existence of different pools of surface-localized GPR183. Chemotaxis mediated by GPR183 depended on receptor desensitization by β-arrestins but could be uncoupled from internalization, highlighting an important biological role for the recruitment of β-arrestin to GPR183. The role of distinct pathways in internalization and chemotaxis may aid in the development of GPR183-targeting drugs for specific disease contexts.
Collapse
Affiliation(s)
- Viktoria M S Kjær
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Viktorija Daugvilaite
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tomasz M Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM)-Pompeu Fabra University (UPF), Barcelona 08003, Spain
- InterAx Biotech AG, Villigen 5234, Switzerland
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw 02-089, Poland
| | - Christian M Madsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid S Jørgensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kaustubh R Bhuskute
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Trond Ulven
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tau Benned-Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Siv A Hjorth
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gertrud M Hjortø
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ee Von Moo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM)-Pompeu Fabra University (UPF), Barcelona 08003, Spain
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Howley K, Berthelette A, Ceglia S, Kang J, Reboldi A. Embryonic type 3 innate lymphoid cells sense maternal dietary cholesterol to control local Peyer's patch development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533339. [PMID: 36993524 PMCID: PMC10055282 DOI: 10.1101/2023.03.19.533339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Lymphoid tissue inducer (LTi) cells develop during intrauterine life and rely on developmental programs to initiate the organogenesis of secondary lymphoid organs (SLOs). This evolutionary conserved process endows the fetus with the ability to orchestrate the immune response after birth and to react to the triggers present in the environment. While it is established that LTi function can be shaped by maternal-derived cues and is critical to prepare the neonate with a functional scaffold to mount immune response, the cellular mechanisms that control anatomically distinct SLO organogenesis remain unclear. We discovered that LTi cells forming Peyer's patches, gut-specific SLOs, require the coordinated action of two migratory G protein coupled receptors (GPCR) GPR183 and CCR6. These two GPCRs are uniformly expressed on LTi cells across SLOs, but their deficiency specifically impacts Peyer's patch formation, even when restricted to fetal window. The unique CCR6 ligand is CCL20, while the ligand for GPR183 is the cholesterol metabolite 7α,25-Dihydroxycholesterol (7α,25-HC), whose production is controlled by the enzyme cholesterol 25-hydroxylase (CH25H). We identified a fetal stromal cell subset that expresses CH25H and attracts LTi cells in the nascent Peyer's patch anlagen. GPR183 ligand concentration can be modulated by the cholesterol content in the maternal diet and impacts LTi cell maturation in vitro and in vivo, highlighting a link between maternal nutrients and intestinal SLO organogenesis. Our findings revealed that in the fetal intestine, cholesterol metabolite sensing by GPR183 in LTi cells for Peyer's patch formation is dominant in the duodenum, the site of cholesterol absorption in the adult. This anatomic requirement suggests that embryonic, long-lived non-hematopoietic cells might exploit adult metabolic functions to ensure highly specialized SLO development in utero.
Collapse
Affiliation(s)
- Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
38
|
Frascoli M, Ferraj E, Miu B, Malin J, Spidale NA, Cowan J, Shissler SC, Brink R, Xu Y, Cyster JG, Bhandoola A, Kang J, Reboldi A. Skin γδ T cell inflammatory responses are hardwired in the thymus by oxysterol sensing via GPR183 and calibrated by dietary cholesterol. Immunity 2023; 56:562-575.e6. [PMID: 36842431 PMCID: PMC10306310 DOI: 10.1016/j.immuni.2023.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/04/2022] [Accepted: 01/24/2023] [Indexed: 02/27/2023]
Abstract
Dietary components and metabolites have a profound impact on immunity and inflammation. Here, we investigated how sensing of cholesterol metabolite oxysterols by γδ T cells impacts their tissue residency and function. We show that dermal IL-17-producing γδ T (Tγδ17) cells essential for skin-barrier homeostasis require oxysterols sensing through G protein receptor 183 (GPR183) for their development and inflammatory responses. Single-cell transcriptomics and murine reporter strains revealed that GPR183 on developing γδ thymocytes is needed for their maturation by sensing medullary thymic epithelial-cell-derived oxysterols. In the skin, basal keratinocytes expressing the oxysterol enzyme cholesterol 25-hydroxylase (CH25H) maintain dermal Tγδ17 cells. Diet-driven increases in oxysterols exacerbate Tγδ17-cell-mediated psoriatic inflammation, dependent on GPR183 on γδ T cells. Hence, cholesterol-derived oxysterols control spatially distinct but biologically linked processes of thymic education and peripheral function of dermal T cells, implicating diet as a focal parameter of dermal Tγδ17 cells.
Collapse
Affiliation(s)
- Michela Frascoli
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Enxhi Ferraj
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Bing Miu
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Justin Malin
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas A Spidale
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jennifer Cowan
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susannah C Shissler
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
39
|
Bai J, Tan BK. B Lineage Cells and IgE in Allergic Rhinitis and CRSwNP and the Role of Omalizumab Treatment. Am J Rhinol Allergy 2023; 37:182-192. [PMID: 36848269 PMCID: PMC10830379 DOI: 10.1177/19458924221147770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) and chronic rhinosinusitis (CRS) are two prevalent nasal diseases where both type 2 inflammation and immunoglobulin E (IgE) may play important roles. Although they can exist independently or comorbidly, subtle but important differences exist in immunopathogenesis. OBJECTIVE To summarize current knowledge of pathophysiological roles of B lineage cells and IgE in AR and CRS with nasal polyps (CRSwNP). METHODS Searched PubMed database, reviewed AR and CRSwNP-related literature, and discussed disease diagnosis, comorbidity, epidemiology, pathophysiology, and treatment. Similarities and differences in B-cell biology and IgE are compared in the 2 conditions. RESULTS Both AR and CRSwNP have evidence for pathological type 2 inflammation, B-cell activation and differentiation, and IgE production. However, distinctions exist in the clinical and serological profiles at diagnosis, as well as treatments utilized. B-cell activation in AR may more frequently be regulated in the germinal center of lymphoid follicles, whereas CRSwNP may occur via extrafollicular pathways although controversies remain in these initial activating events. Oligoclonal and antigen-specific IgE maybe predominate in AR, but polyclonal and antigen-nonspecific IgE may predominate in CRSwNP. Omalizumab has been shown efficacious in treating both AR and CRSwNP in multiple clinical trials but is the only Food and Drug Administration-approved anti-IgE biologic to treat CRSwNP or allergic asthma. Staphylococcus aureus frequently colonizes the nasal airway and has the ability to activate type two responses including B-cell responses although the extent to which it modulates AR and CRSwNP disease severity is being investigated. CONCLUSION This review highlights current knowledge of the roles of B cells and IgE in the pathogenesis of AR and CRSwNP and a small comparison between the 2 diseases. More systemic studies should be done to elevate the understanding of these diseases and their treatment.
Collapse
Affiliation(s)
- Junqin Bai
- Department of Otolaryngology, 12244Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Bruce K Tan
- Department of Otolaryngology, 12244Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Allergy and Immunology, Department of Medicine, 12244Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
40
|
Foo CX, Bartlett S, Chew KY, Ngo MD, Bielefeldt-Ohmann H, Arachchige BJ, Matthews B, Reed S, Wang R, Smith C, Sweet MJ, Burr L, Bisht K, Shatunova S, Sinclair JE, Parry R, Yang Y, Lévesque JP, Khromykh A, Rosenkilde MM, Short KR, Ronacher K. GPR183 antagonism reduces macrophage infiltration in influenza and SARS-CoV-2 infection. Eur Respir J 2023; 61:2201306. [PMID: 36396144 PMCID: PMC9686317 DOI: 10.1183/13993003.01306-2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/20/2022] [Indexed: 11/19/2022]
Abstract
RATIONALE Severe viral respiratory infections are often characterised by extensive myeloid cell infiltration and activation and persistent lung tissue injury. However, the immunological mechanisms driving excessive inflammation in the lung remain poorly understood. OBJECTIVES To identify the mechanisms that drive immune cell recruitment in the lung during viral respiratory infections and identify novel drug targets to reduce inflammation and disease severity. METHODS Preclinical murine models of influenza A virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. RESULTS Oxidised cholesterols and the oxysterol-sensing receptor GPR183 were identified as drivers of monocyte/macrophage infiltration to the lung during influenza A virus (IAV) and SARS-CoV-2 infection. Both IAV and SARS-CoV-2 infection upregulated the enzymes cholesterol 25-hydroxylase (CH25H) and cytochrome P450 family 7 subfamily member B1 (CYP7B1) in the lung, resulting in local production of the oxidised cholesterols 25-hydroxycholesterol (25-OHC) and 7α,25-dihydroxycholesterol (7α,25-OHC). Loss-of-function mutation of Gpr183 or treatment with a GPR183 antagonist reduced macrophage infiltration and inflammatory cytokine production in the lungs of IAV- or SARS-CoV-2-infected mice. The GPR183 antagonist significantly attenuated the severity of SARS-CoV-2 infection and viral loads. Analysis of single-cell RNA-sequencing data on bronchoalveolar lavage samples from healthy controls and COVID-19 patients with moderate and severe disease revealed that CH25H, CYP7B1 and GPR183 are significantly upregulated in macrophages during COVID-19. CONCLUSION This study demonstrates that oxysterols drive inflammation in the lung via GPR183 and provides the first preclinical evidence for the therapeutic benefit of targeting GPR183 during severe viral respiratory infections.
Collapse
Affiliation(s)
- Cheng Xiang Foo
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
- Contributed equally to this work
| | - Stacey Bartlett
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
- Contributed equally to this work
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Minh Dao Ngo
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | | | - Benjamin Matthews
- Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Sarah Reed
- Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Ran Wang
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Christian Smith
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Matthew J Sweet
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Australia
| | - Lucy Burr
- Dept of Respiratory Medicine, Mater Adult Hospital, Brisbane, Australia
| | - Kavita Bisht
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Svetlana Shatunova
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Jane E Sinclair
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Rhys Parry
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Yuanhao Yang
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Jean-Pierre Lévesque
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Alexander Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | | | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - Katharina Ronacher
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
41
|
Conlon TM, Yildirim AÖ. Oxysterol metabolism dictates macrophage influx during SARS-CoV-2 infection. Eur Respir J 2023; 61:13993003.02417-2022. [PMID: 36858446 DOI: 10.1183/13993003.02417-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/07/2023] [Indexed: 03/03/2023]
Affiliation(s)
- Thomas M Conlon
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
42
|
Ceglia S, Berthelette A, Howley K, Li Y, Mortzfeld B, Bhattarai SK, Yiew NKH, Xu Y, Brink R, Cyster JG, Hooper LV, Randolph GJ, Bucci V, Reboldi A. An epithelial cell-derived metabolite tunes immunoglobulin A secretion by gut-resident plasma cells. Nat Immunol 2023; 24:531-544. [PMID: 36658240 PMCID: PMC10243503 DOI: 10.1038/s41590-022-01413-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 12/14/2022] [Indexed: 01/21/2023]
Abstract
Immunoglobulin A (IgA) secretion by plasma cells, terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. Exposure to diet-derived and commensal-derived signals provides immune cells with organizing cues that instruct their effector function and dynamically shape intestinal immune responses at the mucosal barrier. Recent data have described metabolic and microbial inputs controlling T cell and innate lymphoid cell activation in the gut; however, whether IgA-secreting lamina propria plasma cells are tuned by local stimuli is completely unknown. Although antibody secretion is considered to be imprinted during B cell differentiation and therefore largely unaffected by environmental changes, a rapid modulation of IgA levels in response to intestinal fluctuations might be beneficial to the host. In the present study, we showed that dietary cholesterol absorption and commensal recognition by duodenal intestinal epithelial cells lead to the production of oxysterols, evolutionarily conserved lipids with immunomodulatory functions. Using conditional cholesterol 25-hydroxylase deleter mouse line we demonstrated that 7α,25-dihydroxycholesterol from epithelial cells is critical to restrain IgA secretion against commensal- and pathogen-derived antigens in the gut. Intestinal plasma cells sense oxysterols via the chemoattractant receptor GPR183 and couple their tissue positioning with IgA secretion. Our findings revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around plasma cell localization for efficient mucosal protection.
Collapse
Affiliation(s)
- Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yun Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benedikt Mortzfeld
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shakti K Bhattarai
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nicole K H Yiew
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, MO, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University Medical School, St. Louis, MO, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
43
|
Ribeiro ML, Profitós-Pelejà N, Santos JC, Blecua P, Reyes-Garau D, Armengol M, Fernández-Serrano M, Miskin HP, Bosch F, Esteller M, Normant E, Roué G. G protein-coupled receptor 183 mediates the sensitization of Burkitt lymphoma tumors to CD47 immune checkpoint blockade by anti-CD20/PI3Kδi dual therapy. Front Immunol 2023; 14:1130052. [PMID: 37153563 PMCID: PMC10160608 DOI: 10.3389/fimmu.2023.1130052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background Immunotherapy-based regimens have considerably improved the survival rate of B-cell non-Hodgkin lymphoma (B-NHL) patients in the last decades; however, most disease subtypes remain almost incurable. TG-1801, a bispecific antibody that targets CD47 selectively on CD19+ B-cells, is under clinical evaluation in relapsed/refractory (R/R) B-NHL patients either as a single-agent or in combination with ublituximab, a new generation CD20 antibody. Methods A set of eight B-NHL cell lines and primary samples were cultured in vitro in the presence of bone marrow-derived stromal cells, M2-polarized primary macrophages, and primary circulating PBMCs as a source of effector cells. Cell response to TG-1801 alone or combined with the U2 regimen associating ublituximab to the PI3Kδ inhibitor umbralisib, was analyzed by proliferation assay, western blot, transcriptomic analysis (qPCR array and RNA sequencing followed by gene set enrichment analysis) and/or quantification of antibody-dependent cell death (ADCC) and antibody-dependent cell phagocytosis (ADCP). CRISPR-Cas9 gene edition was used to selectively abrogate GPR183 gene expression in B-NHL cells. In vivo, drug efficacy was determined in immunodeficient (NSG mice) or immune-competent (chicken embryo chorioallantoic membrane (CAM)) B-NHL xenograft models. Results Using a panel of B-NHL co-cultures, we show that TG-1801, by disrupting the CD47-SIRPα axis, potentiates anti-CD20-mediated ADCC and ADCP. This led to a remarkable and durable antitumor effect of the triplet therapy composed by TG-1801 and U2 regimen, in vitro, as well as in mice and CAM xenograft models of B-NHL. Transcriptomic analysis also uncovered the upregulation of the G protein-coupled and inflammatory receptor, GPR183, as a crucial event associated with the efficacy of the triplet combination. Genetic depletion and pharmacological inhibition of GPR183 impaired ADCP initiation, cytoskeleton remodeling and cell migration in 2D and 3D spheroid B-NHL co-cultures, and disrupted macrophage-mediated control of tumor growth in B-NHL CAM xenografts. Conclusions Altogether, our results support a crucial role for GPR183 in the recognition and elimination of malignant B cells upon concomitant targeting of CD20, CD47 and PI3Kδ, and warrant further clinical evaluation of this triplet regimen in B-NHL.
Collapse
Affiliation(s)
- Marcelo Lima Ribeiro
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista, São Paulo, Brazil
| | - Núria Profitós-Pelejà
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | | | - Pedro Blecua
- Cancer Epigenetics Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Diana Reyes-Garau
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
| | - Marc Armengol
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
| | - Miranda Fernández-Serrano
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
| | | | - Francesc Bosch
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Hematology, Vall d’Hebron University Hospital, Barcelona, Spain
- Experimental Hematology, Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- Instituciò Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | | - Gael Roué
- Lymphoma Translational Group, Josep Carreras Leukemia Research Institute, Badalona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Hematology, Vall d’Hebron University Hospital, Barcelona, Spain
- Experimental Hematology, Vall d’Hebron Institute of Oncology, Barcelona, Spain
- *Correspondence: Gael Roué,
| |
Collapse
|
44
|
Zheng M, Hu Z, Mei X, Ouyang L, Song Y, Zhou W, Kong Y, Wu R, Rao S, Long H, Shi W, Jing H, Lu S, Wu H, Jia S, Lu Q, Zhao M. Single-cell sequencing shows cellular heterogeneity of cutaneous lesions in lupus erythematosus. Nat Commun 2022; 13:7489. [PMID: 36470882 PMCID: PMC9722937 DOI: 10.1038/s41467-022-35209-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Discoid lupus erythematosus (DLE) and systemic lupus erythematosus (SLE) are both types of lupus, yet the characteristics, and differences between them are not fully understood. Here we show single-cell RNA sequencing data of cutaneous lesions from DLE and SLE patients and skin tissues from healthy controls (HCs). We find significantly higher proportions of T cells, B cells and NK cells in DLE than in SLE. Expanded CCL20+ keratinocyte, CXCL1+ fibroblast, ISGhiCD4/CD8 T cell, ISGhi plasma cell, pDC, and NK subclusters are identified in DLE and SLE compared to HC. In addition, we observe higher cell communication scores between cell types such as fibroblasts and macrophage/dendritic cells in cutaneous lesions of DLE and SLE compared to HC. In summary, we clarify the heterogeneous characteristics in cutaneous lesions between DLE and SLE, and discover some specific cell subtypes and ligand-receptor pairs that indicate possible therapeutic targets of lupus erythematosus.
Collapse
Affiliation(s)
- Meiling Zheng
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Zhi Hu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Xiaole Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Lianlian Ouyang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Yang Song
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Wenhui Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Yi Kong
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Shijia Rao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Hui Jing
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Shuang Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, 410011, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China.
| |
Collapse
|
45
|
Ejam SS, Saleh RO, Catalan Opulencia MJ, Najm MA, Makhmudova A, Jalil AT, Abdelbasset WK, Al-Gazally ME, Hammid AT, Mustafa YF, Sergeevna SE, Karampoor S, Mirzaei R. Pathogenic role of 25-hydroxycholesterol in cancer development and progression. Future Oncol 2022; 18:4415-4442. [PMID: 36651359 DOI: 10.2217/fon-2022-0819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/06/2022] [Indexed: 01/19/2023] Open
Abstract
Cholesterol is an essential lipid that serves several important functions, including maintaining the homeostasis of cells, acting as a precursor to bile acid and steroid hormones and preserving the stability of membrane lipid rafts. 25-hydroxycholesterol (25-HC) is a cholesterol derivative that may be formed from cholesterol. 25-HC is a crucial component in various biological activities, including cholesterol metabolism. In recent years, growing evidence has shown that 25-HC performs a critical function in the etiology of cancer, infectious diseases and autoimmune disorders. This review will summarize the latest findings regarding 25-HC, including its biogenesis, immunomodulatory properties and role in innate/adaptive immunity, inflammation and the development of various types of cancer.
Collapse
Affiliation(s)
| | - Raed Obaid Saleh
- Department of Pharmacy, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Mazin Aa Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Aziza Makhmudova
- Department of Social Sciences & Humanities, Samarkand State Medical Institute, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Walid Kamal Abdelbasset
- Department of Health & Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | | | - Ali Thaeer Hammid
- Computer Engineering Techniques Department, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Sergushina Elena Sergeevna
- National Research Ogarev Mordovia State University, 68 Bolshevitskaya Street, Republic of Mordovia, Saransk, 430005, Russia
| | - Sajad Karampoor
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom & Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
46
|
Braden K, Campolo M, Li Y, Chen Z, Doyle TM, Giancotti LA, Esposito E, Zhang J, Cuzzocrea S, Arnatt CK, Salvemini D. Activation of GPR183 by 7 α,25-Dihydroxycholesterol Induces Behavioral Hypersensitivity through Mitogen-Activated Protein Kinase and Nuclear Factor- κB. J Pharmacol Exp Ther 2022; 383:172-181. [PMID: 36116795 PMCID: PMC9553113 DOI: 10.1124/jpet.122.001283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023] Open
Abstract
Emerging evidence implicates the G-protein coupled receptor (GPCR) GPR183 in the development of neuropathic pain. Further investigation of the signaling pathways downstream of GPR183 is needed to support the development of GPR183 antagonists as analgesics. In rodents, intrathecal injection of its ligand, 7α,25-dihydroxycholesterol (7α,25-OHC), causes time-dependent development of mechano-and cold- allodynia (behavioral hypersensitivity). These effects are blocked by the selective small molecule GPR183 antagonist, SAE-14. However, the molecular mechanisms engaged downstream of GPR183 in the spinal cord are not known. Here, we show that 7α,25-OHC-induced behavioral hypersensitivity is Gα i dependent, but not β-arrestin 2-dependent. Non-biased transcriptomic analyses of dorsal-horn spinal cord (DH-SC) tissues harvested at the time of peak hypersensitivity implicate potential contributions of mitogen-activated protein kinase (MAPK) and nuclear factor κB (NF-κB). In support, we found that the development of 7α,25-OHC/GPR183-induced mechano-allodynia was associated with significant activation of MAPKs (extracellular signal-regulated kinase [ERK], p38) and redox-sensitive transcription factors (NF-κB) and increased formation of inflammatory and neuroexcitatory cytokines. SAE-14 blocked these effects and behavioral hypersensitivity. Our findings provide novel mechanistic insight into how GPR183 signaling in the spinal cord produces hypersensitivity through MAPK and NF-κB activation. SIGNIFICANCE STATEMENT: Using a multi-disciplinary approach, we have characterized the molecular mechanisms underpinning 7α,25-OHC/GPR183-induced hypersensitivity in mice. Intrathecal injections of the GPR183 agonist 7α,25-OHC induce behavioral hypersensitivity, and these effects are blocked by the selective GPR183 antagonist SAE-14. We found that 7α,25-OHC-induced allodynia is dependent on MAPK and NF-κB signaling pathways and results in an increase in pro-inflammatory cytokine expression. This study provides a first insight into how GPR183 signaling in the spinal cord is pronociceptive.
Collapse
Affiliation(s)
- Kathryn Braden
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Michela Campolo
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Ying Li
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Zhoumou Chen
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Luigino Antonio Giancotti
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Emanuela Esposito
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Salvatore Cuzzocrea
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Christopher Kent Arnatt
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| |
Collapse
|
47
|
Dicker M, Li Y, Giles DA, Verstichel G, Castelan VC, Ascui-Gac G, Chou TF, Perez-Jeldres T, Cheroutre H, Kronenberg M. CD4 +-mediated colitis in mice is independent of the GPR183 and GPR18 pathways. Front Immunol 2022; 13:1034648. [PMID: 36389671 PMCID: PMC9652117 DOI: 10.3389/fimmu.2022.1034648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 09/24/2023] Open
Abstract
Colitis is characterized by an exacerbated intestinal immune response, but the genetic and other mechanisms regulating immune activation remain incompletely understood. In order to identify new pathways leading to colitis, we sought to identify genes with increased expression in the colons of patients that also are near loci identified by genome wide association studies (GWAS) associated with IBD risk. One such SNP, rs9557195 was of particular interest because it is within an intron of G-protein-coupled receptor (GPR) 183, known to be important for lymphocyte migration. Furthermore, this SNP is in close proximity to the gene encoding another G-protein coupled receptor, GPR18. Analyzing publicly available datasets, we found transcripts of GPR183 and GPR18 to be increased in colon biopsies from ulcerative colitis and Crohn's disease patients, and GPR183 was even more increased in patients resistant to TNF treatment. Expression of both genes also was increased in mouse models of colitis. Therefore, our aim was to understand if increased expression of these GPRs in the intestine is related to disease severity in colitis models. Here we investigated the role of these receptors in the T cell transfer model and the dextran sulfate sodium model. In the T cell transfer model, GPR183 expression on donor T cells, as well as on other cell types in the Rag-/- recipients, was not essential for severe colitis induction. Furthermore, deficiency in Rag-/- mice for the enzyme that synthesizes a cholesterol metabolite that is a major ligand for GPR183 also did not affect disease. Similarly, lack of GPR18 expression in T cells or other cell types did not affect colitis pathogenesis in the T cell transfer or in the dextran sulfate sodium model. Therefore, despite increased expression of transcripts for these genes in the intestine during inflammation in humans and mice, they are not required for disease severity in mouse models of colitis induced by chemical injury or T cell cytokines, perhaps due to redundancy in mechanisms important for homing and survival of lymphocytes to the inflamed intestine.
Collapse
Affiliation(s)
- Martina Dicker
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Yingcong Li
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA, United States
| | - Daniel A. Giles
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Greet Verstichel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Viankail Cedillo Castelan
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Gabriel Ascui-Gac
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Ting-Fang Chou
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Tamara Perez-Jeldres
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Hilde Cheroutre
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Mitchell Kronenberg
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
48
|
Hasan Z, Nguyen TQ, Lam BWS, Wong JHX, Wong CCY, Tan CKH, Yu J, Thiam CH, Zhang Y, Angeli V, Nguyen LN. Postnatal deletion of Spns2 prevents neuroinflammation without compromising blood vascular functions. Cell Mol Life Sci 2022; 79:541. [PMID: 36198832 PMCID: PMC11802987 DOI: 10.1007/s00018-022-04573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 09/09/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Protein Spinster homolog 2 (Spns2) is a sphingosine-1-phosphate (S1P) transporter that releases S1P to regulate lymphocyte egress and trafficking. Global deletion of Spns2 (Spns2-/-) has been shown to reduce disease severity in several autoimmune disease models. To examine whether Spns2 could be exploited as a drug target, we generated and characterized the mice with postnatal knockout of Spns2 (Spns2-Mx1Cre). Our results showed that Spns2-Mx1Cre mice had significantly low number of lymphocytes in blood and lymphoid organs similar to Spns2-/- mice. Lymph but not plasma S1P levels were significantly reduced in both groups of knockout mice. Our lipidomic results also showed that Spns2 releases different S1P species into lymph. Interestingly, lymphatic vessels in the lymph nodes (LNs) of Spns2-/- and Spns2-Mx1Cre mice exhibited morphological defects. The structures of high endothelial venules (HEV) in the LNs of Spns2-Mx1Cre mice were disorganized. These results indicate that lack of Spns2 affects both S1P secretion and LN vasculatures. Nevertheless, blood vasculature of these Spns2 deficient mice was not different to controls under homeostasis and vascular insults. Importantly, Spns2-Mx1Cre mice were resistant to multiple sclerosis in experimental autoimmune encephalomyelitis (EAE) models with significant reduction of pathogenic Th17 cells in the central nervous system (CNS). This study suggests that pharmacological inhibition of Spns2 may be exploited for therapeutic applications in treatment of neuroinflammation.
Collapse
Affiliation(s)
- Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, Singapore
| | - Jovi Hui Xin Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Caleb Cheng Yi Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Clarissa Kai Hui Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Jiabo Yu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Chung Hwee Thiam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Veronique Angeli
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
- Life Sciences Institute, Singapore Lipidomics Incubator (SLING), National University of Singapore, Singapore, 117456, Singapore.
- Cardiovascular Disease Research (CVD) Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
- Life Sciences Institute, Immunology Program, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
49
|
Zehentmeier S, Lim VY, Ma Y, Fossati J, Ito T, Jiang Y, Tumanov AV, Lee HJ, Dillinger L, Kim J, Csomos K, Walter JE, Choi J, Pereira JP. Dysregulated stem cell niches and altered lymphocyte recirculation cause B and T cell lymphopenia in WHIM syndrome. Sci Immunol 2022; 7:eabo3170. [PMID: 36149943 PMCID: PMC9614684 DOI: 10.1126/sciimmunol.abo3170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gain-of-function (GOF) mutations in CXCR4 cause WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, characterized by infections, leukocyte retention in bone marrow (BM), and blood leukopenias. B lymphopenia is evident at early progenitor stages, yet why do CXCR4 GOF mutations that cause B (and T) lymphopenia remain obscure? Using a CXCR4 R334X GOF mouse model of WHIM syndrome, we showed that lymphopoiesis is reduced because of a dysregulated mesenchymal stem cell (MSC) transcriptome characterized by a switch from an adipogenic to an osteolineage-prone program with limited lymphopoietic activity. We identify lymphotoxin beta receptor (LTβR) as a critical pathway promoting interleukin-7 (IL-7) down-regulation in MSCs. Blocking LTβR or CXCR4 signaling restored IL-7 production and B cell development in WHIM mice. LTβR blocking also increased production of IL-7 and B cell activating factor (BAFF) in secondary lymphoid organs (SLOs), increasing B and T cell numbers in the periphery. These studies revealed that LTβR signaling in BM MSCs and SLO stromal cells limits the lymphocyte compartment size.
Collapse
Affiliation(s)
- Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Vivian Y Lim
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yifan Ma
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Julia Fossati
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yawen Jiang
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ho-Joon Lee
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Lukas Dillinger
- X4 Pharmaceuticals Inc., Cambridge, MA, USA
- X4 Pharmaceuticals Inc., Vienna, Austria
| | - Jihyun Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Krisztian Csomos
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Division Allergy and Immunology, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Jungmin Choi
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| |
Collapse
|
50
|
Frascoli M, Reboldi A, Kang J. Dietary Cholesterol Metabolite Regulation of Tissue Immune Cell Development and Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:645-653. [PMID: 35961669 PMCID: PMC10215006 DOI: 10.4049/jimmunol.2200273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023]
Abstract
Obesity is considered the primary environmental factor associated with morbidity and severity of wide-ranging inflammatory disorders. The molecular mechanism linking high-fat or cholesterol diet to imbalances in immune responses, beyond the increased production of generic inflammatory factors, is just beginning to emerge. Diet cholesterol by-products are now known to regulate function and migration of diverse immune cell subsets in tissues. The hydroxylated metabolites of cholesterol oxysterols as central regulators of immune cell positioning in lymphoid and mucocutaneous tissues is the focus of this review. Dedicated immunocyte cell surface receptors sense spatially distributed oxysterol tissue depots to tune cell metabolism and function, to achieve the "right place at the right time" axiom of efficient tissue immunity.
Collapse
Affiliation(s)
- Michela Frascoli
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|