1
|
Wang Q, Li S, Wei Y, Xu C, Liu X, Wang X, Chai W, Mou W, Chen X, Li C, Wang C, Gui J. An increase in IL-10-producing DNT cells is associated with the pathogenesis of pediatric SLE. Clin Immunol 2025:110490. [PMID: 40158789 DOI: 10.1016/j.clim.2025.110490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that causes immune system overactivity and organ damage. Among T-cell subsets involved in SLE, CD4 and CD8 double-negative αβT (DNT) cells have attracted attention in recent years, although their role in SLE remains poorly understood. Examining the minute intricacies, particularly signaling pathway modifications is crucial, as it may unveil potential therapeutic targets and lead to the development of more effective treatments. Our study found increased DNT cells in pediatric SLE patients, with elevated IL-10 signaling. These IL-10-producing DNT cells were positively related to disease activity defined by SLE Disease Activity Index (SLEDAI), and were further elevated in patients with lupus nephritis. Additionally, our results indicated that IL-10-producing DNT cells correlated positively with anti-Sm autoantibodies. Collectively, our study revealed that modulation of IL-10 production within DNT-cell subset could affect both immune regulation and autoantibody production, contributing to the immunological dysregulation in SLE.
Collapse
Affiliation(s)
- Qixin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Shipeng Li
- Department of Rheumatology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56 Nan Li Shi Lu, Beijing 100045, China
| | - Yannan Wei
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China
| | - Chen Xu
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China
| | - Xiangjun Liu
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xiaolin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wenjia Chai
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xi Chen
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Caifeng Li
- Department of Rheumatology, National Centre for Children's Health, Beijing Children's Hospital, Capital Medical University, No. 56 Nan Li Shi Lu, Beijing 100045, China.
| | - Caisheng Wang
- Inner Mongolia Xilingol League Central Hospital, Xilinhaote 026000, China.
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
2
|
Fajardo-Despaigne JE, Lombard-Vadnais F, Pelletier AN, Olazabal A, Boutin L, Pasquin S, Janelle V, Legault L, Delisle JS, Hillhouse EE, Coderre L, Lesage S. Characterization and effective expansion of CD4 -CD8 - TCRαβ + T cells from individuals living with type 1 diabetes. Mol Ther Methods Clin Dev 2025; 33:101400. [PMID: 39877593 PMCID: PMC11772147 DOI: 10.1016/j.omtm.2024.101400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025]
Abstract
CD4-CD8- TCRαβ+ (double-negative [DN]) T cells represent a rare T cell population that promotes immunological tolerance through various cytotoxic mechanisms. In mice, autologous transfer of DN T cells has shown protective effects against autoimmune diabetes and graft-versus-host disease. Here, we characterized human DN T cells from people living with type 1 diabetes (PWT1D) and healthy controls. We found that while DN T cells and CD8+ T cells share many similarities, DN T cells are a unique T cell population, both at the transcriptomic and protein levels. We also show that by using various cytokine combinations, human DN T cells can be expanded in vitro up to 1,000-fold (mean >250-fold) and remain functional post-expansion. In addition, we report that DN T cells from PWT1D display a phenotype comparable to that of healthy controls, efficiently expand, and are highly functional. As DN T cells are immunoregulatory and can prevent T1D in various mouse models, these observations suggest that autologous DN T cells may be amenable to therapy for the prevention or treatment of T1D.
Collapse
Affiliation(s)
| | - Félix Lombard-Vadnais
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | | | - Aïnhoa Olazabal
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Lucie Boutin
- Département de Recherche Clinique, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC, Canada
| | - Sarah Pasquin
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Valérie Janelle
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Laurent Legault
- Département de Recherche Clinique, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC, Canada
- Department of Pediatrics, Montreal Children’s Hospital, Montreal, QC, Canada
| | - Jean-Sébastien Delisle
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Erin E. Hillhouse
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Lise Coderre
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lesage
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
3
|
Yero A, Shi T, Clain JA, Zghidi-Abouzid O, Racine G, Costiniuk CT, Routy JP, Estaquier J, Jenabian MA. Double-Negative T-Cells during Acute Human Immunodeficiency Virus and Simian Immunodeficiency Virus Infections and Following Early Antiretroviral Therapy Initiation. Viruses 2024; 16:1609. [PMID: 39459942 PMCID: PMC11512404 DOI: 10.3390/v16101609] [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: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
HIV infection significantly affects the frequencies and functions of immunoregulatory CD3+CD4-CD8- double-negative (DN) T-cells, while the effect of early antiretroviral therapy (ART) initiation on these cells remains understudied. DN T-cell subsets were analyzed prospectively in 10 HIV+ individuals during acute infection and following early ART initiation compared to 20 HIV-uninfected controls. In this study, 21 Rhesus macaques (RMs) were SIV-infected, of which 13 were assessed during acute infection and 8 following ART initiation four days post-infection. DN T-cells and FoxP3+ DN Treg frequencies increased during acute HIV infection, which was not restored by ART. The expression of activation (HLA-DR/CD38), immune checkpoints (PD-1/CTLA-4), and senescence (CD28-CD57+) markers by DN T-cells and DN Tregs increased during acute infection and was not normalized by ART. In SIV-infected RMs, DN T-cells remained unchanged despite infection or ART, whereas DN Treg frequencies increased during acute SIV infection and were not restored by ART. Finally, frequencies of CD39+ DN Tregs increased during acute HIV and SIV infections and remained elevated despite ART. Altogether, acute HIV/SIV infections significantly changed DN T-cell and DN Treg frequencies and altered their immune phenotype, while these changes were not fully normalized by early ART, suggesting persistent HIV/SIV-induced immune dysregulation despite early ART initiation.
Collapse
Affiliation(s)
- Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
| | - Tao Shi
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
| | - Julien A. Clain
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Ouafa Zghidi-Abouzid
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Gina Racine
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Cecilia T. Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada; (C.T.C.); (J.-P.R.)
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada; (C.T.C.); (J.-P.R.)
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jérôme Estaquier
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
4
|
Moysidou E, Christodoulou M, Lioulios G, Stai S, Karamitsos T, Dimitroulas T, Fylaktou A, Stangou M. Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis. Int J Mol Sci 2024; 25:10905. [PMID: 39456692 PMCID: PMC11508046 DOI: 10.3390/ijms252010905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by considerable changes in peripheral lymphocyte structure and function, that plays a critical role in commencing and reviving the inflammatory and immune signaling pathways. In healthy individuals, B lymphocytes have a major role in guiding and directing defense mechanisms against pathogens. Certain changes in B lymphocyte phenotype, including alterations in surface and endosomal receptors, occur in the presence of SLE and lead to dysregulation of peripheral B lymphocyte subpopulations. Functional changes are characterized by loss of self-tolerance, intra- and extrafollicular activation, and increased cytokine and autoantibody production. T lymphocytes seem to have a supporting, rather than a leading, role in the disease pathogenesis. Substantial aberrations in peripheral T lymphocyte subsets are evident, and include a reduction of cytotoxic, regulatory, and advanced differentiated subtypes, together with an increase of activated and autoreactive forms and abnormalities in follicular T cells. Up-regulated subpopulations, such as central and effector memory T cells, produce pre-inflammatory cytokines, activate B lymphocytes, and stimulate cell signaling pathways. This review explores the pivotal roles of B and T lymphocytes in the pathogenesis of SLE and Lupus Nephritis, emphasizing the multifaceted mechanisms and interactions and their phenotypic and functional dysregulations.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Theodoros Karamitsos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Cardiology, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros Dimitroulas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 4th Department of Medicine, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
5
|
Rodríguez-Rodríguez N, Rosetti F, Crispín JC. CD8 is down(regulated) for tolerance. Trends Immunol 2024; 45:442-453. [PMID: 38782625 DOI: 10.1016/j.it.2024.04.012] [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: 03/22/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Activated CD8+ T cells directly kill target cells. Therefore, the regulation of their function is central to avoiding immunopathology. Mechanisms that curb effector functions in CD4+ and CD8+ T cells are mostly shared, yet important differences occur. Here, we focus on the control of CD8+ T cell activity and discuss the importance of a poorly understood aspect of tolerance that directly impairs engagement of target cells: the downregulation of CD8. We contextualize this process and propose that it represents a key element during CD8+ T cell modulation.
Collapse
Affiliation(s)
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Mexico.
| |
Collapse
|
6
|
París-Muñoz A, León-Triana O, Pérez-Martínez A, Barber DF. Helios as a Potential Biomarker in Systemic Lupus Erythematosus and New Therapies Based on Immunosuppressive Cells. Int J Mol Sci 2023; 25:452. [PMID: 38203623 PMCID: PMC10778776 DOI: 10.3390/ijms25010452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The Helios protein (encoded by the IKZF2 gene) is a member of the Ikaros transcription family and it has recently been proposed as a promising biomarker for systemic lupus erythematosus (SLE) disease progression in both mouse models and patients. Helios is beginning to be studied extensively for its influence on the T regulatory (Treg) compartment, both CD4+ Tregs and KIR+/Ly49+ CD8+ Tregs, with alterations to the number and function of these cells correlated to the autoimmune phenomenon. This review analyzes the most recent research on Helios expression in relation to the main immune cell populations and its role in SLE immune homeostasis, specifically focusing on the interaction between T cells and tolerogenic dendritic cells (tolDCs). This information could be potentially useful in the design of new therapies, with a particular focus on transfer therapies using immunosuppressive cells. Finally, we will discuss the possibility of using nanotechnology for magnetic targeting to overcome some of the obstacles related to these therapeutic approaches.
Collapse
Affiliation(s)
- Andrés París-Muñoz
- Department of Immunology and Oncology and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain;
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Odelaisy León-Triana
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Domingo F. Barber
- Department of Immunology and Oncology and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain;
| |
Collapse
|
7
|
Paskiewicz A, Niu J, Chang C. Autoimmune lymphoproliferative syndrome: A disorder of immune dysregulation. Autoimmun Rev 2023; 22:103442. [PMID: 37683818 DOI: 10.1016/j.autrev.2023.103442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Autoimmune Lymphoproliferative Syndrome (ALPS) is an autoimmune disease that has been reported in over 2200 patients. It is a rare, genetic disease where pathogenic variants occur in the extrinsic pathway of apoptosis. Various mutations in different genes, such as FAS, FASL, and CASP10, can result in ALPS. Most commonly, pathogenic variants occur in the FAS receptor. This malfunctioning pathway allows for the abnormal accumulation of lymphocytes, namely CD3 + TCRαβ+CD4 - CD8- (double negative (DN) T) cells, which are a hallmark of the disease. This disease usually presents in childhood with lymphadenopathy and splenomegaly as a result of lymphoproliferation. Over time, these patients may develop cytopenias or lymphomas because of irregularities in the immune system. Current treatments include glucocorticoids, mycophenolate mofetil, sirolimus, immunoglobulin G, and rituximab. These medications serve to manage the symptoms and there are no standardized recommendations for the management of ALPS. The only curative therapy is a bone marrow transplant, but this is rarely done because of the complications. This review serves to broaden the understanding of ALPS by discussing the mechanism of immune dysregulation, how the symptoms manifest, and the mechanisms of treatment. Additionally, we discuss the epidemiology, comorbidities, and medications relating to ALPS patients across the United States using data from Cosmos.
Collapse
Affiliation(s)
- Amy Paskiewicz
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA.
| | - Jianli Niu
- Office of Human Research, Memorial Healthcare System, Hollywood, FL 33021, USA.
| | - Christopher Chang
- Division of Immunology, Allergy and Pediatric Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL 33021, USA.
| |
Collapse
|
8
|
Abdelhamid L, Mao J, Cabana-Puig X, Zhu J, Swartwout BK, Edwards MR, Testerman JC, Michaelis JS, Allen IC, Ahmed SA, Luo XM. Nlrp12 deficiency alters gut microbiota and ameliorates Faslpr-mediated systemic autoimmunity in male mice. Front Immunol 2023; 14:1120958. [PMID: 36969209 PMCID: PMC10036793 DOI: 10.3389/fimmu.2023.1120958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
NLRP12 has dual roles in shaping inflammation. We hypothesized that NLRP12 would modulate myeloid cells and T cell function to control systemic autoimmunity. Contrary to our hypothesis, the deficiency of Nlrp12 in autoimmune-prone B6.Faslpr/lpr mice ameliorated autoimmunity in males but not females. Nlrp12 deficiency dampened B cell terminal differentiation, germinal center reaction, and survival of autoreactive B cells leading to decreased production of autoantibodies and reduced renal deposition of IgG and complement C3. In parallel, Nlrp12 deficiency reduced the expansion of potentially pathogenic T cells, including double-negative T cells and T follicular helper cells. Furthermore, reduced pro-inflammatory innate immunity was observed, where the gene deletion decreased in-vivo expansion of splenic macrophages and mitigated ex-vivo responses of bone marrow-derived macrophages and dendritic cells to LPS stimulation. Interestingly, Nlrp12 deficiency altered the diversity and composition of fecal microbiota in both male and female B6/lpr mice. Notably, however, Nlrp12 deficiency significantly modulated small intestinal microbiota only in male mice, suggesting that the sex differences in disease phenotype might be gut microbiota-dependent. Together, these results suggest a potential pathogenic role of NLRP12 in promoting systemic autoimmunity in males. Future studies will investigate sex-based mechanisms through which NLRP12 differentially modulates autoimmune outcomes.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Department of Microbiology, College of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Jiangdi Mao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jing Zhu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Brianna K. Swartwout
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michael R. Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - James C. Testerman
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jacquelyn S. Michaelis
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, United States
| | - Irving Coy Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- *Correspondence: S. Ansar Ahmed, ; Xin M. Luo,
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- *Correspondence: S. Ansar Ahmed, ; Xin M. Luo,
| |
Collapse
|
9
|
Van Meerhaeghe T, Néel A, Brouard S, Degauque N. Regulation of CD8 T cell by B-cells: A narrative review. Front Immunol 2023; 14:1125605. [PMID: 36969196 PMCID: PMC10030846 DOI: 10.3389/fimmu.2023.1125605] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Activation of CD4 T cells by B cells has been extensively studied, but B cell-regulated priming, proliferation, and survival of CD8 T cells remains controversial. B cells express high levels of MHC class I molecules and can potentially act as antigen-presenting cells (APCs) for CD8 T cells. Several in vivo studies in mice and humans demonstrate the role of B cells as modulators of CD8 T cell function in the context of viral infections, autoimmune diseases, cancer and allograft rejection. In addition, B-cell depletion therapies can lead to impaired CD8 T-cell responses. In this review, we attempt to answer 2 important questions: 1. the role of B cell antigen presentation and cytokine production in the regulation of CD8 T cell survival and cell fate determination, and 2. The role of B cells in the formation and maintenance of CD8 T cell memory.
Collapse
Affiliation(s)
- Tess Van Meerhaeghe
- Department of Nephrology, Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
| | - Antoine Néel
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
- Internal Medicine Department, Nantes University Hospital, Nantes, France
| | - Sophie Brouard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
| | - Nicolas Degauque
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
- *Correspondence: Nicolas Degauque,
| |
Collapse
|
10
|
Adoptive Cell Therapy for T-Cell Malignancies. Cancers (Basel) 2022; 15:cancers15010094. [PMID: 36612092 PMCID: PMC9817702 DOI: 10.3390/cancers15010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
T-cell malignancies are often aggressive and associated with poor prognoses. Adoptive cell therapy has recently shown promise as a new line of therapy for patients with hematological malignancies. However, there are currently challenges in applying adoptive cell therapy to T-cell malignancies. Various approaches have been examined in preclinical and clinical studies to overcome these obstacles. This review aims to provide an overview of the recent progress on adoptive cell therapy for T-cell malignancies. The benefits and drawbacks of different types of adoptive cell therapy are discussed. The potential advantages and current applications of innate immune cell-based adoptive cell therapy for T cell malignancies are emphasized.
Collapse
|
11
|
Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic lupus erythematosus: new insights in pathogenesis and therapeutic strategies. J Autoimmun 2022; 132:102870. [PMID: 35872102 DOI: 10.1016/j.jaut.2022.102870] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of immune tolerance and sustained production of autoantibodies. Multiple and profound T cell abnormalities in SLE are intertwined with disease expression. Both numerical and functional disturbances have been reported in main CD4+ T helper cell subsets including Th1, Th2, Th17, regulatory, and follicular helper cells. SLE CD4+ T cells are known to provide help to B cells, produce excessive IL-17 but insufficient IL-2, and infiltrate tissues. In the absence of sufficient amounts of IL-2, regulatory T cells, do not function properly to constrain inflammation. A complicated series of early signaling defects and aberrant activation of kinases and phosphatases result in complex cell phenotypes by altering the metabolic profile and the epigenetic landscape. All main metabolic pathways including glycolysis, glutaminolysis and oxidative phosphorylation are altered in T cells from lupus prone mice and patients with SLE. SLE CD8+ cytotoxic T cells display reduced cytolytic activity which accounts for higher rates of infection and the sustenance of autoimmunity. Further, CD8+ T cells in the context of rheumatic diseases lose the expression of CD8, acquire IL-17+CD4-CD8- double negative T (DNT) cell phenotype and infiltrate tissues. Herein we present an update on these T cell abnormalities along with underlying mechanisms and discuss how these advances can be exploited therapeutically. Novel strategies to correct these aberrations in T cells show promise for SLE treatment.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Afroditi Boulougoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
París-Muñoz A, Aizpurua G, Barber DF. Helios Expression Is Downregulated on CD8+ Treg in Two Mouse Models of Lupus During Disease Progression. Front Immunol 2022; 13:922958. [PMID: 35784310 PMCID: PMC9244697 DOI: 10.3389/fimmu.2022.922958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022] Open
Abstract
T-cell–mediated autoimmunity reflects an imbalance in this compartment that is not restored by tolerogenic immune cells, e.g., regulatory T cells or tolerogenic dendritic cells (tolDCs). Although studies into T-cell equilibrium have mainly focused on regulatory CD4+FoxP3+ T cells (CD4+ Tregs), recent findings on the lesser known CD8+ Tregs (CD44+CD122+Ly49+) have highlighted their non-redundant role in regulating lupus-like disease and their regulatory phenotype facilitated by the transcription factor Helios in mice and humans. However, there are still remaining questions about Helios regulation and dynamics in different autoimmune contexts. Here, we show the absence of CD8+ Tregs in two lupus-prone murine models: MRL/MPJ and MRL/lpr, in comparison with a non-prone mouse strain like C57BL/6. We observed that all MRL animals showed a dramatically reduced population of CD8+ Tregs and a greater Helios downregulation on diseased mice. Helios induction was detected preferentially on CD8+ T cells from OT-I mice co-cultured with tolDCs from C57BL/6 but not in MRL animals. Furthermore, the Helios profile was also altered in other relevant T-cell populations implicated in lupus, such as CD4+ Tregs, conventional CD4+, and double-negative T cells. Together, these findings could make Helios a versatile maker across the T-cell repertoire that is capable of differentiating lupus disease states.
Collapse
Affiliation(s)
- Andrés París-Muñoz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
- NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Gonzalo Aizpurua
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
- NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Domingo F. Barber
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
- NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
- *Correspondence: Domingo F. Barber,
| |
Collapse
|
13
|
Velikkakam T, Gollob KJ, Dutra WO. Double-negative T cells: Setting the stage for disease control or progression. Immunology 2022; 165:371-385. [PMID: 34939192 PMCID: PMC10626195 DOI: 10.1111/imm.13441] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022] Open
Abstract
Double-negative (DN) T cells are present at relatively low frequencies in human peripheral blood, and are characterized as expressing the alpha-beta or gamma-delta T-cell receptor (TCR), but not the CD4 nor the CD8 co-receptors. Despite their low frequencies, these cells are potent producers of cytokines and, thus, are key orchestrators of immune responses. DN T cells were initially associated with induction of peripheral immunological tolerance and immunomodulatory activities related to disease prevention. However, other studies demonstrated that these cells can also display effector functions associated with pathology development. This apparent contradiction highlighted the heterogeneity of the DN T-cell population. Here, we review phenotypic and functional characteristics of DN T cells, emphasizing their role in human diseases. The need for developing biomarkers to facilitate the translation of studies from animal models to humans will also be discussed. Finally, we will examine DN T cells as promising therapeutic targets to prevent or inhibit human disease development.
Collapse
Affiliation(s)
- Teresiama Velikkakam
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einsten, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais – INCT-DT, Belo Horizonte, Brazil
| | - Walderez Ornelas Dutra
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Pós-graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais – INCT-DT, Belo Horizonte, Brazil
| |
Collapse
|
14
|
Regulation of activated T cell survival in rheumatic autoimmune diseases. Nat Rev Rheumatol 2022; 18:232-244. [PMID: 35075294 DOI: 10.1038/s41584-021-00741-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/29/2022]
Abstract
Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.
Collapse
|
15
|
Barcenilla H, Pihl M, Wahlberg J, Ludvigsson J, Casas R. Intralymphatic GAD-alum Injection Modulates B Cell Response and Induces Follicular Helper T Cells and PD-1+ CD8+ T Cells in Patients With Recent-Onset Type 1 Diabetes. Front Immunol 2022; 12:797172. [PMID: 35095874 PMCID: PMC8791064 DOI: 10.3389/fimmu.2021.797172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022] Open
Abstract
Antigen-specific immunotherapy is an appealing strategy to preserve beta-cell function in type 1 diabetes, although the approach has yet to meet its therapeutic endpoint. Direct administration of autoantigen into lymph nodes has emerged as an alternative administration route that can improve the efficacy of the treatment. In the first open-label clinical trial in humans, injection of aluminum-formulated glutamic acid decarboxylase (GAD-alum) into an inguinal lymph node led to the promising preservation of C-peptide in patients with recent-onset type 1 diabetes. The treatment induced a distinct immunomodulatory effect, but the response at the cell level has not been fully characterized. Here we used mass cytometry to profile the immune landscape in peripheral blood mononuclear cells from 12 participants of the study before and after 15 months of treatment. The immunomodulatory effect of the therapy included reduction of naïve and unswitched memory B cells, increase in follicular helper T cells and expansion of PD-1+ CD69+ cells in both CD8+ and double negative T cells. In vitro stimulation with GAD65 only affected effector CD8+ T cells in samples collected before the treatment. However, the recall response to antigen after 15 months included induction of CXCR3+ and CD11c+Tbet+ B cells, PD-1+ follicular helper T cells and exhausted-like CD8+ T cells. This study provides a deeper insight into the immunological changes associated with GAD-alum administration directly into the lymph nodes.
Collapse
Affiliation(s)
- Hugo Barcenilla
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Mikael Pihl
- Core Facility, Flow Cytometry Unit, Linköping University, Linköping, Sweden
| | - Jeanette Wahlberg
- Department of Health, Medicine and Caring Sciences (HMV), Linköping University, Linköping, Sweden.,Division of Diagnostics and Specialist Medicine and Faculty of Health Sciences, Örebro University, Örebro, Sweden
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden.,Division of Pediatrics, Crown Princess Victoria Children's Hospital, Linköping, Sweden
| | - Rosaura Casas
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
16
|
Péter B, Boldizsár I, Kovács GM, Erdei A, Bajtay Z, Vörös A, Ramsden JJ, Szabó I, Bősze S, Horvath R. Natural Compounds as Target Biomolecules in Cellular Adhesion and Migration: From Biomolecular Stimulation to Label-Free Discovery and Bioactivity-Based Isolation. Biomedicines 2021; 9:1781. [PMID: 34944597 PMCID: PMC8698624 DOI: 10.3390/biomedicines9121781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023] Open
Abstract
Plants and fungi can be used for medical applications because of their accumulation of special bioactive metabolites. These substances might be beneficial to human health, exerting also anti-inflammatory and anticancer (antiproliferative) effects. We propose that they are mediated by influencing cellular adhesion and migration via various signaling pathways and by directly inactivating key cell adhesion surface receptor sites. The evidence for this proposition is reviewed (by summarizing the natural metabolites and their effects influencing cellular adhesion and migration), along with the classical measuring techniques used to gain such evidence. We systematize existing knowledge concerning the mechanisms of how natural metabolites affect adhesion and movement, and their role in gene expression as well. We conclude by highlighting the possibilities to screen natural compounds faster and more easily by applying new label-free methods, which also enable a far greater degree of quantification than the conventional methods used hitherto. We have systematically classified recent studies regarding the effects of natural compounds on cellular adhesion and movement, characterizing the active substances according to their organismal origin (plants, animals or fungi). Finally, we also summarize the results of recent studies and experiments on SARS-CoV-2 treatments by natural extracts affecting mainly the adhesion and entry of the virus.
Collapse
Affiliation(s)
- Beatrix Péter
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| | - Imre Boldizsár
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary; (I.B.); (G.M.K.)
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Gábor M. Kovács
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary; (I.B.); (G.M.K.)
- Plant Protection Institute, Centre for Agricultural Research, Hungarian Academy of Sciences, 1022 Budapest, Hungary
| | - Anna Erdei
- Department of Immunology, Eötvös Loránd University, 1117 Budapest, Hungary; (A.E.); (Z.B.)
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary
| | - Zsuzsa Bajtay
- Department of Immunology, Eötvös Loránd University, 1117 Budapest, Hungary; (A.E.); (Z.B.)
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, 1117 Budapest, Hungary
| | - Alexandra Vörös
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| | - Jeremy J. Ramsden
- Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, UK;
| | - Ildikó Szabó
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (I.S.); (S.B.)
- National Public Health Center, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - Szilvia Bősze
- MTA-ELTE Research Group of Peptide Chemistry, Eötvös Loránd Research Network (ELKH), Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (I.S.); (S.B.)
- National Public Health Center, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Group, Research Centre for Energy Research, Institute for Technical Physics and Materials Science, Konkoly-Thege u 29-33, 1120 Budapest, Hungary; (A.V.); (R.H.)
| |
Collapse
|
17
|
Jamaly S, Rakaee M, Abdi R, Tsokos GC, Fenton KA. Interplay of immune and kidney resident cells in the formation of tertiary lymphoid structures in lupus nephritis. Autoimmun Rev 2021; 20:102980. [PMID: 34718163 DOI: 10.1016/j.autrev.2021.102980] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/31/2021] [Indexed: 02/07/2023]
Abstract
Kidney involvement confers significant morbidity and mortality in patients with systemic lupus erythematosus (SLE). The pathogenesis of lupus nephritis (LN) involves diverse mechanisms instigated by elements of the autoimmune response which alter the biology of kidney resident cells. Processes in the glomeruli and in the interstitium may proceed independently albeit crosstalk between the two is inevitable. Podocytes, mesangial cells, tubular epithelial cells, kidney resident macrophages and stromal cells with input from cytokines and autoantibodies present in the circulation alter the expression of enzymes, produce cytokines and chemokines which lead to their injury and damage of the kidney. Several of these molecules can be targeted independently to prevent and reverse kidney failure. Tertiary lymphoid structures with true germinal centers are present in the kidneys of patients with lupus nephritis and have been increasingly recognized to associate with poorer renal outcomes. Stromal cells, tubular epithelial cells, high endothelial vessel and lymphatic venule cells produce chemokines which enable the formation of structures composed of a T-cell-rich zone with mature dendritic cells next to a B-cell follicle with the characteristics of a germinal center surrounded by plasma cells. Following an overview on the interaction of the immune cells with kidney resident cells, we discuss the cellular and molecular events which lead to the formation of tertiary lymphoid structures in the interstitium of the kidneys of mice and patients with lupus nephritis. In parallel, molecules and processes that can be targeted therapeutically are presented.
Collapse
Affiliation(s)
- Simin Jamaly
- Department of Medical Biology, Faculty of Health Science, UiT The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Mehrdad Rakaee
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reza Abdi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kristin Andreassen Fenton
- Department of Medical Biology, Faculty of Health Science, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a serious autoimmune disease with a wide range of organ involvement. In addition to aberrant B-cell responses leading to autoantibody production, T-cell abnormalities are important in the induction of autoimmunity and the ensuing downstream organ damage. In this article, we present an update on how subsets of CD8+ T cells contribute to SLE pathogenesis. RECENT FINDINGS Reduced cytolytic function of CD8+ T cells not only promotes systemic autoimmunity but also accounts for the increased risk of infections. Additional information suggests that effector functions of tissue CD8+ T cells contribute to organ damage. The phenotypic changes in tissue CD8+ T cells likely arise from exposure to tissue microenvironment and crosstalk with tissue resident cells. Research on pathogenic IL-17-producing double negative T cells also suggests their origin from autoreactive CD8+ T cells, which also contribute to the induction and maintenance of systemic autoimmunity. SUMMARY Reduced CD8+ T-cell effector function illustrates their role in peripheral tolerance in the control of autoimmunity and to the increased risk of infections. Inflammatory cytokine producing double negative T cells and functional defects of regulatory CD8+ T cell both contribute to SLE pathogenesis. Further in depth research on these phenotypic changes are warranted for the development of new therapeutics for people with SLE.
Collapse
|
19
|
Flores-Mendoza G, Rodríguez-Rodríguez N, Rubio RM, Madera-Salcedo IK, Rosetti F, Crispín JC. Fas/FasL Signaling Regulates CD8 Expression During Exposure to Self-Antigens. Front Immunol 2021; 12:635862. [PMID: 33841416 PMCID: PMC8024570 DOI: 10.3389/fimmu.2021.635862] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/04/2021] [Indexed: 11/13/2022] Open
Abstract
Activation of self-reactive CD8+ T cells induces a peripheral tolerance mechanism that involves loss of CD8 expression. Because genetic deficiency of Fas and Fasl causes the accumulation of double-negative (DN; CD3+ TCR-αβ+ CD4- CD8-) T cells that have been proposed to derive from CD8+ cells, we decided to explore the role of Fas and FasL in self-antigen-induced CD8 downregulation. To this end, we quantified Fas and FasL induction by different stimuli and analyzed the effects of Fas/FasL deficiency during a protective immune response and after exposure to self-antigens. Our data describes how Fas and FasL upregulation differs depending on the setting of CD8 T cell activation and demonstrates that Fas/FasL signaling maintains CD8 expression during repetitive antigen stimulation and following self-antigen encounter. Together, our results reveal an unexpected role of Fas/FasL signaling and offer a new insight into the role of these molecules in the regulation of immune tolerance.
Collapse
Affiliation(s)
- Giovanna Flores-Mendoza
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Noé Rodríguez-Rodríguez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rosa M. Rubio
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Iris K. Madera-Salcedo
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Florencia Rosetti
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C. Crispín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Mexico
| |
Collapse
|
20
|
Sharma J, Collins TD, Roach T, Mishra S, Lam BK, Mohamed ZS, Veal AE, Polk TB, Jones A, Cornaby C, Haider MI, Zeumer-Spataro L, Johnson HM, Morel LM, Larkin J. Suppressor of cytokine signaling-1 mimetic peptides attenuate lymphocyte activation in the MRL/lpr mouse autoimmune model. Sci Rep 2021; 11:6354. [PMID: 33737712 PMCID: PMC7973732 DOI: 10.1038/s41598-021-86017-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/09/2021] [Indexed: 12/30/2022] Open
Abstract
Autoimmune diseases are driven largely by a pathogenic cytokine milieu produced by aberrantly activated lymphocytes. Many cytokines, including interferon gamma (IFN-γ), utilize the JAK/STAT pathway for signal propagation. Suppressor of Cytokine Signaling-1 (SOCS1) is an inducible, intracellular protein that regulates IFN-γ signaling by dampening JAK/STAT signaling. Using Fas deficient, MRL/MpJ-Faslpr/J (MRL/lpr) mice, which develop lupus-like disease spontaneously, we tested the hypothesis that a peptide mimic of the SOCS1 kinase inhibitory region (SOCS1-KIR) would inhibit lymphocyte activation and modulate lupus-associated pathologies. Consistent with in vitro studies, SOCS1-KIR intraperitoneal administration reduced the frequency, activation, and cytokine production of memory CD8+ and CD4+ T lymphocytes within the peripheral blood, spleen, and lymph nodes. In addition, SOCS1-KIR administration reduced lymphadenopathy, severity of skin lesions, autoantibody production, and modestly reduced kidney pathology. On a cellular level, peritoneal SOCS1-KIR administration enhanced Foxp3 expression in total splenic and follicular regulatory T cells, reduced the effector memory/naïve T lymphocyte ratio for both CD4+ and CD8+ cells, and reduced the frequency of GL7+ germinal center enriched B cells. Together, these data show that SOCS1-KIR treatment reduced auto-reactive lymphocyte effector functions and suggest that therapeutic targeting of the SOCS1 pathway through peptide administration may have efficacy in mitigating autoimmune pathologies.
Collapse
Affiliation(s)
- Jatin Sharma
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Teresa D Collins
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Tracoyia Roach
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Shiwangi Mishra
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Brandon K Lam
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Zaynab Sidi Mohamed
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Antia E Veal
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Timothy B Polk
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Amari Jones
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Caleb Cornaby
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mohammed I Haider
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Leilani Zeumer-Spataro
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Howard M Johnson
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA
| | - Laurence M Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Joseph Larkin
- Department of Microbiology & Cell Science, University of Florida, Museum Road Building 981, PO Box 110700, Gainesville, FL, 32611, USA.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW TCRαβ+CD4-CD8- double-negative T (DNT) cells, a principal subset of mature T lymphocytes, have been closely linked with autoimmune/inflammatory conditions. However, controversy persists regarding their ontogeny and function. Here, we present an overview on DNT cells in different autoimmune diseases to advance a deeper understanding of the contribution of this population to disease pathogenesis. RECENT FINDINGS DNT cells have been characterized in various chronic inflammatory diseases and they have been proposed to display pathogenic or regulatory function. The tissue location of DNT cells and the effector cytokines they produce bespeak to their active involvement in chronic inflammatory diseases. SUMMARY By producing various cytokines, expanded DNT cells in inflamed tissues contribute to the pathogenesis of a variety of autoimmune inflammatory diseases. However, it is unclear whether this population represents a stable lineage consisting of different subsets similar to CD4+ T helper cell subset. Better understanding of the possible heterogeneity and plasticity of DNT cells is needed to reveal interventional therapeutic opportunities.
Collapse
Affiliation(s)
- Hao Li
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
22
|
HIV Infection and Persistence in Pulmonary Mucosal Double Negative T Cells In Vivo. J Virol 2020; 94:JVI.01788-20. [PMID: 32967958 PMCID: PMC7925170 DOI: 10.1128/jvi.01788-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 11/20/2022] Open
Abstract
The lungs are relatively unexplored anatomical human immunodeficiency virus (HIV) reservoirs in the antiretroviral therapy (ART) era. Double negative (DN) T cells are a subset of T cells that lack expression of CD4 and CD8 (CD4- CD8-) and may have both regulatory and effector functions during HIV infection. Notably, circulating DN T cells were previously described as cellular HIV reservoirs. Here, we undertook a thorough analysis of pulmonary versus blood DN T cells of people living with HIV (PLWH) under ART. Bronchoalveolar lavage (BAL) fluid and matched peripheral blood were collected from 35 PLWH on ART and 16 uninfected volunteers without respiratory symptoms. Both PLWH and HIV-negative (HIV-) adults displayed higher frequencies of DN T cells in BAL versus blood, and these cells mostly exhibited an effector memory phenotype. In PLWH, pulmonary mucosal DN T cells expressed higher levels of HLA-DR and several cellular markers associated with HIV persistence (CCR6, CXCR3, and PD-1) than blood. We also observed that DN T cells were less senescent (CD28- CD57+) and expressed less immunosuppressive ectonucleotidase (CD73/CD39), granzyme B, and perforin in the BAL fluid than in the blood of PLWH. Importantly, fluorescence-activated cell sorter (FACS)-sorted DN T cells from the BAL fluid of PLWH under suppressive ART harbored HIV DNA. Using the humanized bone marrow-liver-thymus (hu-BLT) mouse model of HIV infection, we observed higher infection frequencies of lung DN T cells than those of the blood and spleen in both early and late HIV infection. Overall, our findings show that HIV is seeded in pulmonary mucosal DN T cells early following infection and persists in these potential cellular HIV reservoirs even during long-term ART.IMPORTANCE Reservoirs of HIV during ART are the primary reasons why HIV/AIDS remains an incurable disease. Indeed, HIV remains latent and unreachable by antiretrovirals in cellular and anatomical sanctuaries, preventing its eradication. The lungs have received very little attention compared to other anatomical reservoirs despite being immunological effector sites exhibiting characteristics ideal for HIV persistence. Furthermore, PLWH suffer from a high burden of pulmonary non-opportunistic infections, suggesting impaired pulmonary immunity despite ART. Meanwhile, various immune cell populations have been proposed to be cellular reservoirs in blood, including CD4- CD8- DN T cells, a subset that may originate from CD4 downregulation by HIV proteins. The present study aims to describe DN T cells in human and humanized mice lungs in relation to intrapulmonary HIV burden. The characterization of DN T cells as cellular HIV reservoirs and the lungs as an anatomical HIV reservoir will contribute to the development of targeted HIV eradication strategies.
Collapse
|
23
|
Moroney JB, Vasudev A, Pertsemlidis A, Zan H, Casali P. Integrative transcriptome and chromatin landscape analysis reveals distinct epigenetic regulations in human memory B cells. Nat Commun 2020; 11:5435. [PMID: 33116135 PMCID: PMC7595102 DOI: 10.1038/s41467-020-19242-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023] Open
Abstract
Memory B cells (MBCs) are long-lived and produce high-affinity, generally, class-switched antibodies. Here, we use a multiparameter approach involving CD27 to segregate naïve B cells (NBC), IgD+ unswitched (unsw)MBCs and IgG+ or IgA+ class-switched (sw)MBCs from humans of different age, sex and race. Conserved antibody variable gene expression indicates that MBCs emerge through unbiased selection from NBCs. Integrative analyses of mRNAs, miRNAs, lncRNAs, chromatin accessibility and cis-regulatory elements uncover a core mRNA-ncRNA transcriptional signature shared by IgG+ and IgA+ swMBCs and distinct from NBCs, while unswMBCs display a transitional transcriptome. Some swMBC transcriptional signature loci are accessible but not expressed in NBCs. Profiling miRNAs reveals downregulated MIR181, and concomitantly upregulated MIR181 target genes such as RASSF6, TOX, TRERF1, TRPV3 and RORα, in swMBCs. Finally, lncRNAs differentially expressed in swMBCs cluster proximal to the IgH chain locus on chromosome 14. Our findings thus provide new insights into MBC transcriptional programs and epigenetic regulation, opening new investigative avenues on these critical cell elements in human health and disease. Human memory B cells differentiate from naïve B cells and can express different immunoglobulin (Ig) isotypes resulted from class-switch recombination. Here the authors describe, using transcriptional and epigenetic data from human memory B cells and integrated multi-omics analyses, the differentiation regulation and trajectory of IgG+, IgA+ and IgD+ memory B cells.
Collapse
Affiliation(s)
- Justin B Moroney
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Anusha Vasudev
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Hong Zan
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA
| | - Paolo Casali
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
24
|
Li H, Adamopoulos IE, Moulton VR, Stillman IE, Herbert Z, Moon JJ, Sharabi A, Krishfield S, Tsokos MG, Tsokos GC. Systemic lupus erythematosus favors the generation of IL-17 producing double negative T cells. Nat Commun 2020; 11:2859. [PMID: 32503973 PMCID: PMC7275084 DOI: 10.1038/s41467-020-16636-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/08/2020] [Indexed: 01/06/2023] Open
Abstract
Mature double negative (DN) T cells are a population of αβ T cells that lack CD4 and CD8 coreceptors and contribute to systemic lupus erythematosus (SLE). The splenic marginal zone macrophages (MZMs) are important for establishing immune tolerance, and loss of their number or function contributes to the progression of SLE. Here we show that loss of MZMs impairs the tolerogenic clearance of apoptotic cells and alters the serum cytokine profile, which in turn provokes the generation of DN T cells from self-reactive CD8+ T cells. Increased Ki67 expression, narrowed TCR V-beta repertoire usage and diluted T-cell receptor excision circles confirm that DN T cells from lupus-prone mice and patients with SLE undergo clonal proliferation and expansion in a self-antigen dependent manner, which supports the shared mechanisms for their generation. Collectively, our results provide a link between the loss of MZMs and the expansion of DN T cells, and indicate possible strategies to prevent the development of SLE.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Iannis E Adamopoulos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95817, USA
| | - Vaishali R Moulton
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Isaac E Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Zach Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, 21-27 Burlington Ave, Boston, MA, 02215, USA
| | - James J Moon
- Center for Immunology and inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Amir Sharabi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Suzanne Krishfield
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Fortner KA, Blanco LP, Buskiewicz I, Huang N, Gibson PC, Cook DL, Pedersen HL, Yuen PST, Murphy MP, Perl A, Kaplan MJ, Budd RC. Targeting mitochondrial oxidative stress with MitoQ reduces NET formation and kidney disease in lupus-prone MRL- lpr mice. Lupus Sci Med 2020; 7:e000387. [PMID: 32343673 PMCID: PMC7199895 DOI: 10.1136/lupus-2020-000387] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Recent investigations in humans and mouse models with lupus have revealed evidence of mitochondrial dysfunction and production of mitochondrial reactive oxygen species (mROS) in T cells and neutrophils. This can provoke numerous cellular changes including oxidation of nucleic acids, proteins, lipids and even induction of cell death. We have previously observed that in T cells from patients with lupus, the increased mROS is capable of provoking oligomerisation of mitochondrial antiviral stimulator (MAVS) and production of type I interferon (IFN-I). mROS in SLE neutrophils also promotes the formation of neutrophil extracellular traps (NETs), which are increased in lupus and implicated in renal damage. As a result, in addition to traditional immunosuppression, more comprehensive treatments for lupus may also include non-immune therapy, such as antioxidants. METHODS Lupus-prone MRL-lpr mice were treated from weaning for 11 weeks with the mitochondria-targeted antioxidant, MitoQ (200 µM) in drinking water. Mice were then assessed for ROS production in neutrophils, NET formation, MAVS oligomerisation, serum IFN-I, autoantibody production and renal function. RESULTS MitoQ-treated mice manifested reduced neutrophil ROS and NET formation, decreased MAVS oligomerisation and serum IFN-I, and reduced immune complex formation in kidneys, despite no change in serum autoantibody . CONCLUSIONS These findings reveal the potential utility of targeting mROS in addition to traditional immunosuppressive therapy for lupus.
Collapse
Affiliation(s)
- Karen A Fortner
- Vermont Center for Immunology and Infectious Diseases, Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Luz P Blanco
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Iwona Buskiewicz
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY, New York
| | - Nick Huang
- Rheumatology Clinic, Upstate University Hospital, Syracuse, NY, New York
| | - Pamela C Gibson
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Deborah L Cook
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Hege L Pedersen
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutic Unit, Kidney Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, United States
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge, CB2 0XY, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Andras Perl
- Rheumatology Clinic, Upstate University Hospital, Syracuse, NY, New York
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ralph C Budd
- Vermont Center for Immunology and Infectious Diseases, Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
26
|
Rodríguez-Rodríguez N, Flores-Mendoza G, Apostolidis SA, Rosetti F, Tsokos GC, Crispín JC. TCR-α/β CD4 - CD8 - double negative T cells arise from CD8 + T cells. J Leukoc Biol 2020; 108:851-857. [PMID: 32052478 DOI: 10.1002/jlb.1ab0120-548r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 01/02/2023] Open
Abstract
The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/β+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/β+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Current address: Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Giovanna Flores-Mendoza
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Current address: Division of Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - José C Crispín
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, Mexico
| |
Collapse
|
27
|
Abstract
Clinical success attained in patients with cancer treated with checkpoint inhibitors has renewed the interest in the immune system and in particular in T cells as a therapeutic tool to eliminate tumors. Here, we discuss recent studies that evaluate the anti-tumor role of CD8 T cells and the mechanisms that interfere with this function. In particular, we review recent literature that has reported on the phenotype and transcriptome of tumor-infiltrating CD8 T cells and deciphered the mechanisms associated with failed tumor rejection.
Collapse
Affiliation(s)
- José C Crispin
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Mexico City, Mexico.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, USA.,Harvard Medical School, Boston, USA
| |
Collapse
|
28
|
Smith EMD, Lythgoe H, Midgley A, Beresford MW, Hedrich CM. Juvenile-onset systemic lupus erythematosus: Update on clinical presentation, pathophysiology and treatment options. Clin Immunol 2019; 209:108274. [PMID: 31678365 DOI: 10.1016/j.clim.2019.108274] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 12/25/2022]
Abstract
Juvenile-onset systemic lupus erythematosus (jSLE) accounts for up to 20% of all SLE patients. Key differences between juvenile- and adult-onset (aSLE) disease include higher disease activity, earlier development of damage, and increased use of immunosuppressive treatment in jSLE suggesting (at least partial) infectivity secondary to variable pathomechanisms. While the exact pathophysiology of jSLE remains unclear, genetic factors, immune complex deposition, complement activation, hormonal factors and immune cell dysregulation are involved to variable extents, promising future patient stratification based on immune phenotypes. Though less effective and potentially toxic, jSLE patients are treated based upon evidence from studies in aSLE cohorts. Here, age-specific clinical features of jSLE, underlying pathomechanisms, treatment options and disease outcomes will be addressed. Future directions to improve the care of jSLE patients, including implementation of the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) recommendations, biomarkers, treat to target and personalized medicine approaches are discussed.
Collapse
Affiliation(s)
- Eve Mary Dorothy Smith
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| | - Hanna Lythgoe
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Angela Midgley
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK
| | - Michael William Beresford
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Christian Michael Hedrich
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| |
Collapse
|
29
|
Suarez-Fueyo A, Tsokos MG, Kwok SK, Maeda K, Katsuyama E, Lapchak PH, Tsokos GC. Hyaluronic Acid Synthesis Contributes to Tissue Damage in Systemic Lupus Erythematosus. Front Immunol 2019; 10:2172. [PMID: 31572382 PMCID: PMC6753633 DOI: 10.3389/fimmu.2019.02172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
Hyaluronic acid (HA), a component of the extracellular matrix, is the ligand for CD44 and has been implicated in the pathogenesis of kidney inflammation in patients with systemic lupus erythematosus (SLE), but its direct role and mechanism of action have not been studied. Here we show that administration of hymecromone (4-Methylumbelliferone, 4-MU), an HA synthesis inhibitor, to lupus-prone mice suppressed dramatically lupus-related pathology. Interestingly, 4-MU stopped the appearance of disease when administered prior to its onset and inhibited the progression of disease when administered after its appearance. Inhibition of HA synthesis in vivo reduced tissue damage and the number of intrarenal lymphoid cell infiltrates including double negative CD3+CD4-CD8- T cells which are known to be involved in the pathogenesis of SLE. Exposure of human peripheral blood mononuclear cells to HA in vitro increased the generation of CD3+CD4-CD8- T cells through a mechanism involving Rho-associated kinase. Our results signify the importance of the HA-rich tissue microenvironment in the activation of lymphocytes to cause tissue damage in SLE and suggest the consideration of inhibition of HA synthesis to treat patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Akiyama C, Tsumiyama K, Uchimura C, Honda E, Miyazaki Y, Sakurai K, Miura Y, Hashiramoto A, Felsher DW, Shiozawa S. Conditional Upregulation of IFN-α Alone Is Sufficient to Induce Systemic Lupus Erythematosus. THE JOURNAL OF IMMUNOLOGY 2019; 203:835-843. [PMID: 31324723 DOI: 10.4049/jimmunol.1801617] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/17/2019] [Indexed: 11/19/2022]
Abstract
The cause of systemic lupus erythematosus (SLE) is unknown. IFN-α has been suggested as a causative agent of SLE; however, it was not proven, and to what extent and how IFN-α contributes to the disease is unknown. We studied the contribution of IFN-α to SLE by generating inducible IFN-α transgenic mice and directly show that conditional upregulation of IFN-α alone induces a typical manifestation of SLE in the mice not prone to autoimmunity, such as serum immune complex, autoantibody against dsDNA (anti-dsDNA Ab), and the organ manifestations classical to SLE, such as immune complex-deposited glomerulonephritis, classical splenic onion-skin lesion, alopecia, epidermal liquefaction, and positive lupus band test of the skin. In the spleen of mice, activated effector CD4 T cells, IFN-γ-producing CD8 T cells, B220+CD86+ cells, and CD11c+CD86+ cells were increased, and the T cells produced increased amounts of IL-4, IL-6, IL-17, and IFN-γ and decreased IL-2. In particular, activated CD3+CD4-CD8- double-negative T cells positive for TCRαβ, B220, CD1d-teteramer, PD-1, and Helios (that produced increased amounts of IFN-γ, IL-4, IL-17, and TNF-α) were significantly expanded. They infiltrated into kidney and induced de novo glomerulonephritis and alopecia when transferred into naive recipients. Thus, sole upregulation of IFN-α is sufficient to induce SLE, and the double-negative T cells expanded by IFN-α are directly responsible for the organ manifestations, such as lupus skin disease or nephritis.
Collapse
Affiliation(s)
- Chieri Akiyama
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Ken Tsumiyama
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan.,Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| | - Chiaki Uchimura
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Eriko Honda
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Yumi Miyazaki
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Keiichi Sakurai
- Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| | - Yasushi Miura
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Akira Hashiramoto
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Dean W Felsher
- Division of Oncology, Department of Medicine and Pathology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Shunichi Shiozawa
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan; .,Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| |
Collapse
|
31
|
Crispin JC, Hedrich CM, Suárez-Fueyo A, Comte D, Tsokos GC. SLE-Associated Defects Promote Altered T Cell Function. Crit Rev Immunol 2019; 37:39-58. [PMID: 29431078 DOI: 10.1615/critrevimmunol.2018025213] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease linked to profound defects in the function and phenotype of T lymphocytes. Here, we describe abnormal signaling pathways that have been documented in T cells from patients with SLE and discuss how they impact gene expression and immune function, in order to understand how they contribute to disease development and progression.
Collapse
Affiliation(s)
- Jose C Crispin
- Departamento de Inmunologia y Reumatologia, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Christian M Hedrich
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Abel Suárez-Fueyo
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Denis Comte
- Divisions of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - George C Tsokos
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Miller S, Tsou PS, Coit P, Gensterblum-Miller E, Renauer P, Rohraff DM, Kilian NC, Schonfeld M, Sawalha AH. Hypomethylation of STAT1 and HLA-DRB1 is associated with type-I interferon-dependent HLA-DRB1 expression in lupus CD8+ T cells. Ann Rheum Dis 2019; 78:519-528. [PMID: 30674474 PMCID: PMC6679955 DOI: 10.1136/annrheumdis-2018-214323] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 01/14/2023]
Abstract
OBJECTIVE We examined genome-wide DNA methylation changes in CD8+ T cells from patients with lupus and controls and investigated the functional relevance of some of these changes in lupus. METHODS Genome-wide DNA methylation of lupus and age, sex and ethnicity-matched control CD8+ T cells was measured using the Infinium MethylationEPIC arrays. Measurement of relevant cell subsets was performed via flow cytometry. Gene expression was quantified by qPCR. Inhibiting STAT1 and CIITA was performed using fludarabine and CIITA siRNA, respectively. RESULTS Lupus CD8+ T cells had 188 hypomethylated CpG sites compared with healthy matched controls. Among the most hypomethylated were sites associated with HLA-DRB1. Genes involved in the type-I interferon response, including STAT1, were also found to be hypomethylated. IFNα upregulated HLA-DRB1 expression on lupus but not control CD8+ T cells. Lupus and control CD8+ T cells significantly increased STAT1 mRNA levels after treatment with IFNα. The expression of CIITA, a key interferon/STAT1 dependent MHC-class II regulator, is induced by IFNα in lupus CD8+ T cells, but not healthy controls. CIITA knockdown and STAT1 inhibition experiments revealed that HLA-DRB1 expression in lupus CD8+ T cells is dependent on CIITA and STAT1 signalling. Coincubation of naïve CD4+ T cells with IFNα-treated CD8+ T cells led to CD4+ T cell activation, determined by increased expression of CD69 and cytokine production, in patients with lupus but not in healthy controls. This can be blocked by neutralising antibodies targeting HLA-DR. CONCLUSIONS Lupus CD8+ T cells are epigenetically primed to respond to type-I interferon. We describe an HLA-DRB1+ CD8+ T cell subset that can be induced by IFNα in patients with lupus. A possible pathogenic role for CD8+ T cells in lupus that is dependent on a high type-I interferon environment and epigenetic priming warrants further characterisation.
Collapse
Affiliation(s)
- Shaylynn Miller
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick Coit
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Paul Renauer
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dallas M Rohraff
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nathan C Kilian
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark Schonfeld
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
TSOKOS GEORGEC, TSOKOS MARIAG. TARGETING TARGETED TREATMENT FOR IMMUNE AND NON-IMMUNE KIDNEY DISEASES. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2019; 130:88-99. [PMID: 31516171 PMCID: PMC6735968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We have found that calcium calmodulin kinase IV is increased in T cells, podocytes, and mesangial cells from patients with systemic lupus erythematosus, as well as in lupus-prone mice, podocytes of patients with focal segmental glomerulosclerosis, and in mice injected with doxorubicin. We showed that this accounts for aberrant T cell function and glomerular damage. Using nanoparticles (nlg) loaded with a small drug inhibitor of calcium calmodulin kinase IV and tagged with antibodies directed to CD4 we have been able to show inhibition of autoimmunity and lupus nephritis. Also, using nlg tagged with antibodies to nephrin, we showed suppression of nephritis in lupus-prone mice and of glomerular damage in mice exposed to doxorubicin. We propose the development of approaches to deliver drugs to cells in a targeted and precise manner.
Collapse
Affiliation(s)
- GEORGE C. TSOKOS
- Correspondence and reprint requests: George C. Tsokos, MD, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School,
330 Brookline Ave., CLS 937, Boston, Massachusetts 02215617-735-4161
| | | |
Collapse
|
34
|
Li H, Tsokos MG, Bickerton S, Sharabi A, Li Y, Moulton VR, Kong P, Fahmy TM, Tsokos GC. Precision DNA demethylation ameliorates disease in lupus-prone mice. JCI Insight 2018; 3:120880. [PMID: 30135300 DOI: 10.1172/jci.insight.120880] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022] Open
Abstract
Defective DNA methylation in T cells leads to a series of T cell abnormalities in lupus; however, the full effect of T cell lineage-specific DNA methylation on disease expression has not been explored. Here, we show that 5-azacytidine, a DNA methyltransferase inhibitor, targeted to either CD4 or CD8 T cells in mice with established disease using a nanolipogel delivery system dramatically ameliorates lupus-related pathology through distinct mechanisms. In vivo targeted delivery of 5-azacytidine into CD4 T cells favors the expansion and function of Foxp3+ Tregs, whereas targeted delivery to CD8 T cells enhances the cytotoxicity and restrains the expansion of pathogenic TCR-αβ+CD4-CD8- double-negative T cells. Our results signify the importance of cell-specific inhibition of DNA methylation in the treatment of established lupus.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Amir Sharabi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Vaishali R Moulton
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Tarek M Fahmy
- Department of Biomedical Engineering.,Department of Immunobiology, and.,Department of Chemical and Environmental Engineering, Yale University School of Medicine, New Haven, Connecticut, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Hedrich CM, Smith EMD, Beresford MW. Juvenile-onset systemic lupus erythematosus (jSLE) - Pathophysiological concepts and treatment options. Best Pract Res Clin Rheumatol 2018; 31:488-504. [PMID: 29773269 DOI: 10.1016/j.berh.2018.02.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The systemic autoimmune/inflammatory condition systemic lupus erythematosus (SLE) manifests before the age of 16 years in 10-20% of all cases. Clinical courses are more severe, and organ complications are more common in patients with juvenile SLE. Varying gender distribution in different age groups and increasing severity with younger age and the presence of monogenic disease in early childhood indicate distinct differences in the pathophysiology of juvenile versus adult-onset SLE. Regardless of these differences, classification criteria and treatment options are identical. In this article, we discuss age-specific pathomechanisms of juvenile-onset SLE, which are currently available and as future treatment options, and propose reclassification of different forms of SLE along the inflammatory spectrum from autoinflammation to autoimmunity.
Collapse
Affiliation(s)
- Christian M Hedrich
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| | - Eve M D Smith
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Michael W Beresford
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|
36
|
|
37
|
|
38
|
Moulton VR, Suarez-Fueyo A, Meidan E, Li H, Mizui M, Tsokos GC. Pathogenesis of Human Systemic Lupus Erythematosus: A Cellular Perspective. Trends Mol Med 2017. [PMID: 28623084 DOI: 10.1016/j.molmed.2017.05.006] [Citation(s) in RCA: 297] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease affecting multiple organs. A complex interaction of genetics, environment, and hormones leads to immune dysregulation and breakdown of tolerance to self-antigens, resulting in autoantibody production, inflammation, and destruction of end-organs. Emerging evidence on the role of these factors has increased our knowledge of this complex disease, guiding therapeutic strategies and identifying putative biomarkers. Recent findings include the characterization of genetic/epigenetic factors linked to SLE, as well as cellular effectors. Novel observations have provided an improved understanding of the contribution of tissue-specific factors and associated damage, T and B lymphocytes, as well as innate immune cell subsets and their corresponding abnormalities. The intricate web of involved factors and pathways dictates the adoption of tailored therapeutic approaches to conquer this disease.
Collapse
Affiliation(s)
- Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Abel Suarez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Esra Meidan
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Rheumatology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Li
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Hedrich CM, Mäbert K, Rauen T, Tsokos GC. DNA methylation in systemic lupus erythematosus. Epigenomics 2017; 9:505-525. [PMID: 27885845 PMCID: PMC6040049 DOI: 10.2217/epi-2016-0096] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/12/2016] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease facilitated by aberrant immune responses directed against cells and tissues, resulting in inflammation and organ damage. In the majority of patients, genetic predisposition is accompanied by additional factors conferring disease expression. While the exact molecular mechanisms remain elusive, epigenetic alterations in immune cells have been demonstrated to play a key role in disease pathogenesis through the dysregulation of gene expression. Since epigenetic marks are dynamic, allowing cells and tissues to differentiate and adjust, they can be influenced by environmental factors and also be targeted in therapeutic interventions. Here, we summarize reports on DNA methylation patterns in SLE, underlying molecular defects and their effect on immune cell function. We discuss the potential of DNA methylation as biomarker or therapeutic target in SLE.
Collapse
Affiliation(s)
- Christian M Hedrich
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katrin Mäbert
- Pediatric Rheumatology & Immunology, Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Rauen
- Department of Nephrology & Clinical Immunology, RWTH University Hospital, Aachen, Germany
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Suárez-Fueyo A, Bradley SJ, Klatzmann D, Tsokos GC. T cells and autoimmune kidney disease. Nat Rev Nephrol 2017; 13:329-343. [PMID: 28287110 DOI: 10.1038/nrneph.2017.34] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glomerulonephritis is traditionally considered to result from the invasion of the kidney by autoantibodies and immune complexes from the circulation or following their formation in situ, and by cells of the innate and the adaptive immune system. The inflammatory response leads to the proliferation and dysfunction of cells of the glomerulus, and invasion of the interstitial space with immune cells, resulting in tubular cell malfunction and fibrosis. T cells are critical drivers of autoimmunity and related organ damage, by supporting B-cell differentiation and antibody production or by directly promoting inflammation and cytotoxicity against kidney resident cells. T cells might become activated by autoantigens in the periphery and become polarized to secrete inflammatory cytokines before entering the kidney where they have the opportunity to expand owing to the presence of costimulatory molecules and activating cytokines. Alternatively, naive T cells could enter the kidney where they become activated after encountering autoantigen and expand locally. As not all individuals with a peripheral autoimmune response to kidney antigens develop glomerulonephritis, the contribution of local kidney factors expressed or produced by kidney cells is probably of crucial importance. Improved understanding of the biochemistry and molecular biology of T cells in patients with glomerulonephritis offers unique opportunities for the recognition of treatment targets for autoimmune kidney disease.
Collapse
Affiliation(s)
- Abel Suárez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| | - Sean J Bradley
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| | - David Klatzmann
- Sorbonne Universités, Pierre and Marie Curie University, INSERM UMR_S 959, 83 Boulevard de l'Hôpital, F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Clinical Investigation Center in Biotherapy and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), 83 boulevard de l'Hôpital, F-75013, Paris, France
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, CLS-937, Boston, Massachusetts 02215, USA
| |
Collapse
|
41
|
Balomenos D, Shokri R, Daszkiewicz L, Vázquez-Mateo C, Martínez-A C. On How Fas Apoptosis-Independent Pathways Drive T Cell Hyperproliferation and Lymphadenopathy in lpr Mice. Front Immunol 2017; 8:237. [PMID: 28344578 PMCID: PMC5344898 DOI: 10.3389/fimmu.2017.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/20/2017] [Indexed: 01/20/2023] Open
Abstract
Fas induces massive apoptosis in T cells after repeated in vitro T cell receptor (TCR) stimulation and is critical for lymphocyte homeostasis in Fas-deficient (lpr) mice. Although the in vitro Fas apoptotic mechanism has been defined, there is a large conceptual gap between this in vitro phenomenon and the pathway that leads to in vivo development of lymphadenopathy and autoimmunity. A striking abnormality in lpr mice is the excessive proliferation of CD4+ and CD8+ T cells, and more so of the double-negative TCR+CD4−CD8−B220+ T cells. The basis of lpr T cell hyperproliferation remains elusive, as it cannot be explained by Fas-deficient apoptosis. T cell-directed p21 overexpression reduces hyperactivation/hyperproliferation of all lpr T cell subtypes and lymphadenopathy in lpr mice. p21 controls expansion of repeatedly stimulated T cells without affecting apoptosis. These results confirm a direct link between hyperactivation/hyperproliferation, autoreactivity, and lymphadenopathy in lpr mice and, with earlier studies, suggest that Fas apoptosis-independent pathways control lpr T cell hyperproliferation. lpr T cell hyperproliferation could be an indirect result of the defective apoptosis of repeatedly stimulated lpr T cells. Nonetheless, in this perspective, we argue for an alternative setting, in which lack of Fas would directly cause lpr T cell hyperactivation/hyperproliferation in vivo. We propose that Fas/Fas ligand (FasL) acts as an activation inhibitor of recurrently stimulated T cells, and that its disruption causes overexpansion of T cells in lpr mice. Research to define the underlying mechanism of this Fas/FasL effect could resolve the phenotype of lpr mice and lead to therapeutics for related human syndromes.
Collapse
Affiliation(s)
- Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Rahman Shokri
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| |
Collapse
|
42
|
Potential role of IL-17-producing CD4/CD8 double negative αβ T cells in psoriatic skin inflammation in a TPA-induced STAT3C transgenic mouse model. J Dermatol Sci 2016; 85:27-35. [PMID: 27810232 DOI: 10.1016/j.jdermsci.2016.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 10/03/2016] [Accepted: 10/17/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Psoriasis is one of the most common immune-mediated chronic inflammatory skin disorders and is accompanied by erythematous scaly plaques. There is growing evidence that the IL-23/Th17 axis plays a critical role in development of the disease. It was recently shown that in addition to CD4+ Th17 cells, various IL-17-producing cell subsets such as CD8+ Tc17 cells, dermal γδ T cells, and innate lymphoid cells are also involved in the development of psoriatic inflammation in humans. OBJECTIVE To investigate which subsets of IL-17-producing cells are involved in psoriasis-like skin inflammation in a TPA (tumor promoter 12-O-tetradecanoylphorbol-13-acetate)-induced K14.Stat3C mouse model. METHOD Skin-infiltrating cells were isolated from inflamed lesions of TPA-treated K14.Stat3C transgenic mice, and analyzed for IL-17 producing cell subsets by flow cytometry. RESULTS We observed significantly increased numbers of IL-17-producing CD4+ T cells, CD8+ T cells and dermal γδ T cells in TPA-induced skin lesions of K14.Stat3C mice. Additionally, we found that another IL-17-producing T cell subset, αβ-TCR+ CD4CD8 double negative T cells (DN αβ T cells), was also increased in lesional skin. These IL-17-producing DN αβ T cells are NK1.1 negative, suggesting they are not natural killer T cells or mucosal associated invariant T cells. As well as other IL-17-producing cells, DN αβ T cells in the inflamed skin can also respond to IL-23 stimulation to produce IL-17. It is also suggested that DN αβ T cells may express retinoic acid-related orphan receptor gamma t and CC chemokine receptor 6. CONCLUSION In TPA-induced lesional skin of K14.Stat3C mice, IL-17-producing CD4+ Th17 cells, CD8+ Tc17 cells, dermal γδ T cells and TCR- cells probably containing ILCs all participated in skin inflammation, which is similar to human clinical psoriatic features. Furthermore, we showed for the first time the possibility that an IL-17-producing DN αβ T cell subset is also involved in psoriatic inflammation.
Collapse
|
43
|
Suárez-Fueyo A, Bradley SJ, Tsokos GC. T cells in Systemic Lupus Erythematosus. Curr Opin Immunol 2016; 43:32-38. [PMID: 27636649 DOI: 10.1016/j.coi.2016.09.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/28/2016] [Accepted: 09/02/2016] [Indexed: 11/27/2022]
Abstract
Systemic Lupus Erythematosus is an autoimmune disorder caused by a complex combination of genetic, epigenetic and environmental factors. Different polymorphisms and epigenetic modifications lead to altered gene expression and function of several molecules which lead to abnormal T cell responses. Metabolic and functional alterations result in peripheral tolerance failures and biased differentiation of T cells into pro-inflammatory and B cell-helper phenotypes as well as the accumulation of disease-promoting memory T cells. Understanding these T cell alterations and their origins is necessary to develop more accurate patient classification systems and to discover new therapeutic targets.
Collapse
Affiliation(s)
- Abel Suárez-Fueyo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Sean J Bradley
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Wang J, Mizui M, Zeng LF, Bronson R, Finnell M, Terhorst C, Kyttaris VC, Tsokos GC, Zhang ZY, Kontaridis MI. Inhibition of SHP2 ameliorates the pathogenesis of systemic lupus erythematosus. J Clin Invest 2016; 126:2077-92. [PMID: 27183387 DOI: 10.1172/jci87037] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/20/2016] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a devastating multisystemic autoimmune disorder. However, the molecular mechanisms underlying its pathogenesis remain elusive. Some patients with Noonan syndrome, a congenital disorder predominantly caused by gain-of-function mutations in the protein tyrosine phosphatase SH2 domain-containing PTP (SHP2), have been shown to develop SLE, suggesting a functional correlation between phosphatase activity and systemic autoimmunity. To test this directly, we measured SHP2 activity in spleen lysates isolated from lupus-prone MRL/lpr mice and found it was markedly increased compared with that in control mice. Similar increases in SHP2 activity were seen in peripheral blood mononuclear cells isolated from lupus patients relative to healthy patients. To determine whether SHP2 alters autoimmunity and related immunopathology, we treated MRL/lpr mice with an SHP2 inhibitor and found increased life span, suppressed crescentic glomerulonephritis, reduced spleen size, and diminished skin lesions. SHP2 inhibition also reduced numbers of double-negative T cells, normalized ERK/MAPK signaling, and decreased production of IFN-γ and IL-17A/F, 2 cytokines involved in SLE-associated organ damage. Moreover, in cultured human lupus T cells, SHP2 inhibition reduced proliferation and decreased production of IFN-γ and IL-17A/F, further implicating SHP2 in lupus-associated immunopathology. Taken together, these data identify SHP2 as a critical regulator of SLE pathogenesis and suggest targeting of its activity as a potent treatment for lupus patients.
Collapse
|
45
|
Rodríguez-Rodríguez N, Apostolidis SA, Fitzgerald L, Meehan BS, Corbett AJ, Martín-Villa JM, McCluskey J, Tsokos GC, Crispín JC. Pro-inflammatory self-reactive T cells are found within murine TCR-αβ(+) CD4(-) CD8(-) PD-1(+) cells. Eur J Immunol 2016; 46:1383-91. [PMID: 27060346 DOI: 10.1002/eji.201546056] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/23/2016] [Accepted: 03/30/2016] [Indexed: 11/07/2022]
Abstract
TCR-αβ(+) double negative (DN) T cells (CD3(+) TCR-αβ(+) CD4(-) CD8(-) NK1.1(-) CD49b(-) ) represent a minor heterogeneous population in healthy humans and mice. These cells have been ascribed pro-inflammatory and regulatory capacities and are known to expand during the course of several autoimmune diseases. Importantly, previous studies have shown that self-reactive CD8(+) T cells become DN after activation by self-antigens, suggesting that self-reactive T cells may exist within the DN T-cell population. Here, we demonstrate that programmed cell death 1 (PD-1) expression in unmanipulated mice identifies a subset of DN T cells with expression of activation-associated markers and a phenotype that strongly suggests they are derived from self-reactive CD8(+) cells. We also found that, within DN T cells, the PD-1(+) subset generates the majority of pro-inflammatory cytokines. Finally, using a TCR-activation reporter mouse (Nur77-GFP), we confirmed that in the steady-state PD-1(+) DN T cells engage endogenous antigens in healthy mice. In conclusion, we provide evidence that indicates that the PD-1(+) fraction of DN T cells represents self-reactive cells.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren Fitzgerald
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bronwyn S Meehan
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Alexandra J Corbett
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - José Manuel Martín-Villa
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - James McCluskey
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - José C Crispín
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
46
|
Wiener A, Schippers A, Wagner N, Tacke F, Ostendorf T, Honke N, Tenbrock K, Ohl K. CXCR5 is critically involved in progression of lupus through regulation of B cell and double-negative T cell trafficking. Clin Exp Immunol 2016; 185:22-32. [PMID: 26990531 DOI: 10.1111/cei.12791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 02/12/2016] [Accepted: 02/19/2016] [Indexed: 11/28/2022] Open
Abstract
The recruitment of immune cells to sites of tissue inflammation is orchestrated by chemokine/chemokine receptor networks. Among these, the CXCL13/CXCR5 axis is thought to be involved critically in systemic lupus erythematosus (SLE) and lupus nephritis pathogenesis. Beyond B cell abnormalities, another hallmark of SLE disease is the occurrence of aberrant T cell responses. In particular, double-negative (DN) T cells are expanded in the peripheral blood of patients with SLE and in lupus-prone mice. DN T cells induce immunoglobulin production, secrete proinflammatory cytokines and infiltrate inflamed tissue, including kidneys. We aimed to investigate how CXCR5 deficiency changes immune cell trafficking in murine lupus. We therefore crossed CXCR5(-/-) mice with B6/lpr mice, a well-established murine lupus model. B cell numbers and B cellular immune responses were diminished in CXCR5-deficient B6/lpr mice. In addition, we observed reduced accumulation of DN T cells in spleen and lymph nodes, paralleled by reduced splenomegaly and lymphadenopathy. In-vivo migration assays revealed reduced migration of CXCR5-deficient DN T cells into lymph nodes, and ex-vivo-activated CXCR5-deficient DN T cells failed to infiltrate kidneys of recipients. Moreover, DN T cells and B cells of CXCR5-deficient B6/lpr mice failed to migrate towards CXCL13 in vitro. We propose that CXCR5 is involved critically in B cell trafficking and germinal cell (GC) formation in murine lupus and in guiding pathogenic DN T cells into lymphoid organs and kidneys, and we therefore describe new pathomechanisms for the CXCL13/CXCR5 axis in SLE.
Collapse
Affiliation(s)
| | | | | | - F Tacke
- Department of Internal Medicine III
| | - T Ostendorf
- Department of Nephrology and Clinical Immunology, Medical Faculty, RWTH, Aachen, Germany
| | | | | | | |
Collapse
|