1
|
Jiang Z, Ruan X, Zhou X, Li S, Wang C, Huang L, He Z, Zhang Y, Wen C. Phlorizin attenuates lupus nephritis via upregulating PI3K/Akt pathway-mediated Treg differentiation. Int Immunopharmacol 2025; 154:114607. [PMID: 40186900 DOI: 10.1016/j.intimp.2025.114607] [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: 08/26/2024] [Revised: 03/03/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Lupus nephritis (LN) leads to widespread kidney damage and nephron loss, establishing it as a major contributor to acute and chronic kidney injury, which can progress to end-stage renal disease. Phlorizin (PHZ), a major pharmacologically active constituent derived from Lithocarpus polystachyus Rehd., has been shown to exhibit significant immunomodulatory and anti-inflammatory properties. Growing evidence indicates that PHZ may exert a protective influence on kidney function. However, the therapeutic effect and mechanism of PHZ in treating LN need to be elucidated. METHODS The PHZ-associated targets were identified through tools such as PharmMapper, SwissTargetPrediction, SuperPred and Targetnet. Simultaneously, LN-associated target spots were retrieved fromOMIM, DisGeNET, GeneCards, and GEO databases. Additionally, Venny 2.1.0 was employed to analyze the overlap between drug targets and disease targets. Following this, the DAVID software was employed to perform enrichment analyses for GO terms and KEGG pathways on the shared drug-disease target sites. Following this, the construction of protein-protein interaction (PPI) networks for these intersecting targets was carried out using the STRING database and Cytoscape software, aiming to pinpoint critical targets. Ultimately, molecular docking alongside dynamic simulations was used to evaluate the binding affinity between PHZ and the critical genes. Based on these findings, PHZ or Dexamethasone (DXSM) was administered to female MRL/lpr mice, which are predisposed to lupus. The therapeutic effects of PHZ on LN were evaluated by assessing renal function and the degree of kidney inflammation. Concurrently, flow cytometry was employed to measure the percentage of CD4+ T cell subsets. Additionally, relevant signaling pathways were examined through western blot analysis. Furthermore, CD4+CD25+Foxp3+ regulatory T (Treg) cells were induced in vitro. Flow cytometry and immunoblotting were performed to confirm the role and mechanism of PHZ in Treg cell differentiation. RESULTS The PHZ compound specifically targeted 161 genes associated with LN. PPI analysis revealed that among all the target genes, Akt1, ALB, MMP9, HSP90AA1, and NF-κB1 exhibited the highest centrality. KEGG pathway analysis suggested that the phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT) signaling pathway could play a crucial role in the treatment of LN. Molecular docking revealed that PHZ exhibits a strong affinity for binding with AKT1. Experimental studies, both in vitro and in vivo, showed that PHZ might alleviate LN by promoting Treg differentiation via activation of the PI3K/AKT signaling pathway. CONCLUSIONS Integrating network pharmacology, bioinformatics, and experimental validation, our study systematically deciphers the therapeutic efficacy and molecular mechanisms of PHZ against LN. Network pharmacology analysis and bioinformatics suggested PI3K/AKT signaling as the pivotal pathway to treat LN, while subsequent in vivo and in vitro experiments confirmed that PHZ exerts its therapeutic effects through activating the PI3K/AKT signaling pathway, ultimately driving FOXP3-dependent regulatory T cell differentiation.
Collapse
Affiliation(s)
- Zhangsheng Jiang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinyi Ruan
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xingchen Zhou
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Suling Li
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenxi Wang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lin Huang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhixing He
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yun Zhang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chengping Wen
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
2
|
Lyu MA, Tang X, Raso MG, Huang M, Zeng K, Sadeghi T, Flowers CR, Parmar S. Ruxolitinib synergizes with regulatory T cells to improve inflammation but has no added benefits in decreasing albuminuria in SLE. Front Immunol 2025; 16:1449693. [PMID: 39975551 PMCID: PMC11836023 DOI: 10.3389/fimmu.2025.1449693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025] Open
Abstract
Background Umbilical cord blood (UCB)-derived CD4+CD25+CD127low regulatory T cells (Tregs) can decrease albuminuria and anti-dsDNA IgG in systemic lupus erythematosus (SLE). Ruxolitinib, a JAK/STAT inhibitor, has been shown to improve cutaneous manifestations of SLE. We hypothesize that the addition of ruxolitinib to UCB-Tregs may improve SLE outcomes. Methods In vitro cell suppression, phenotype change, IL-10 secretion, and cytokine levels in coculture supernatants were determined to quantify the impact of adding ruxolitinib to UCB-Tregs. A xenogeneic SLE model was utilized to study their in vivo combination. Results In a dose-dependent manner, ruxolitinib addition synergizes with UCB-Tregs to suppress SLE-PBMC proliferation, inhibit CD8+ T cells, and reduce phosphorylation of STAT3/STAT5/AKT in CD8+ T cells. UCB-Treg and ruxolitinib combination also downregulates the soluble form of inflammatory cytokines including IFN-γ, IP-10, TNF-α, IL-6, sCD40L, IL-17A, IL-17F, IL-1α, and LIF in cocultures. The addition of ruxolitinib increases UCB-Treg cell persistence in peripheral blood in vivo and decreases the soluble form of human inflammatory cytokines including IFN-γ, TNF-α, and sCD40L in plasma along with improvement of skin lesions in SLE xenografts. Compared to control, significantly lesser CD3+, CD4+, CD8+, and Ki-67+ infiltrates are observed in the lung and kidney of UCB-Tregs and/or ruxolitinib recipients. No added benefit of addition of ruxolitinib is observed on the significant improvement in the urine albumin/creatinine ratio and the anti-dsDNA IgG levels induced by UCB-Tregs. Conclusions Our results demonstrate that the addition of ruxolitinib to UCB-Tregs increases UCB-Tregs suppressor function and their persistence in vivo, downregulates systemic inflammation, and controls cutaneous SLE but does not add to UCB-Treg-mediated improvement in renal manifestations.
Collapse
Affiliation(s)
- Mi-Ae Lyu
- Department of Lymphoma/Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Ximing Tang
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Meixian Huang
- Department of Lymphoma/Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Ke Zeng
- Department of Lymphoma/Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | | | - Christopher R. Flowers
- Department of Lymphoma/Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Simrit Parmar
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
3
|
Honing DY, Luiten RM, Matos TR. Regulatory T Cell Dysfunction in Autoimmune Diseases. Int J Mol Sci 2024; 25:7171. [PMID: 39000278 PMCID: PMC11241405 DOI: 10.3390/ijms25137171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Regulatory T cells (Tregs), a suppressive subpopulation of T cells, are potent mediators of peripheral tolerance, responsible for immune homeostasis. Many autoimmune diseases exhibit disruptions in Treg function or quantity, resulting in an imbalance between protective and pathogenic immune cells. Selective expansion or manipulation of Tregs is a promising therapeutic approach for autoimmune diseases. However, the extensive diversity of Treg subpopulations and the multiple approaches used for Treg identification leads to high complexity, making it difficult to develop a successful treatment capable of modulating Tregs. In this review, we describe the suppressive mechanisms, subpopulations, classification, and identification methodology for Tregs, and their role in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Dionne Y Honing
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Rosalie M Luiten
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Tiago R Matos
- Department of Dermatology, Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Sanofi, 1105 BP Amsterdam, The Netherlands
| |
Collapse
|
4
|
Papachristodoulou E, Kyttaris VC. New and emerging therapies for systemic lupus erythematosus. Clin Immunol 2024; 263:110200. [PMID: 38582250 DOI: 10.1016/j.clim.2024.110200] [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: 02/10/2024] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Systemic Lupus Erythematosus (SLE) and lupus nephritis treatment is still based on non-specific immune suppression despite the first biological therapy for the disease having been approved more than a decade ago. Intense basic and translational research has uncovered a multitude of pathways that are actively being evaluated as treatment targets in SLE and lupus nephritis, with two new medications receiving FDA approval in the last 3 years. Herein we provide an overview of targeted therapies for SLE including medications targeting the B lymphocyte compartment, intracellular signaling, co-stimulation, and finally the interferons and other cytokines.
Collapse
Affiliation(s)
- Eleni Papachristodoulou
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vasileios C Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Mizui M, Kono M. Novel therapeutic strategies targeting abnormal T-cell signaling in systemic lupus erythematosus. Clin Immunol 2024; 262:110182. [PMID: 38458302 DOI: 10.1016/j.clim.2024.110182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Therapeutic strategies for autoimmune diseases have been based on the use of glucocorticoids and immunosuppressive agents that broadly suppress immune responses. Therefore, organ damage from long-term use and infections due to immunocompromised status have been significant issues. Safer immunosuppressants and biological agents are now available, but there is still an urgent need to develop specific drugs to replace glucocorticoids. T-lymphocytes, central players in immune responses, could be crucial targets in the treatment of autoimmune diseases. Extensive research has been conducted on the phenotypic changes of T-cells in systemic lupus erythematosus, which has led to the discovery of various therapeutic strategies. In this comprehensive review, we discuss novel treatment approaches and target molecules with expected effectiveness in humans and mice, based on research for lymphocytes involved in autoimmune diseases, especially T-cells in SLE.
Collapse
Affiliation(s)
- Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Michihito Kono
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
6
|
Lorenzon R, Ribet C, Pitoiset F, Aractingi S, Banneville B, Beaugerie L, Berenbaum F, Cacoub P, Champey J, Chazouilleres O, Corpechot C, Fautrel B, Mekinian A, Regnier E, Saadoun D, Salem JE, Sellam J, Seksik P, Vicaut E, Rosenzwajg M, Klatzmann D. The universal effects of low-dose interleukin-2 across 13 autoimmune diseases in a basket clinical trial. J Autoimmun 2024; 144:103172. [PMID: 38330545 DOI: 10.1016/j.jaut.2024.103172] [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: 11/06/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND A Tregs insufficiency is central to autoimmune and inflammatory diseases pathophysiology and low dose interleukin-2 (IL-2LD) can specifically activate Tregs. OBJECTIVE To assess IL-2LD therapeutic potential and select diseases for further clinical development, we performed an open-label, phase 2a, disease-finding, "basket trial" involving patients with one of 13 different autoimmune diseases. METHODS 81 patients treated with IL-2LD (1 million IU/day) for 5 days, followed by fortnightly injections. The first 48 patients received diluted Proleukin®, while the subsequent 33 received ready-to-use ILT-101®. The primary endpoint was the change in Tregs at day-8 compared to baseline. Key secondary endpoints included clinical efficacy assessments using the Clinical Global Impression (CGI) scale, disease-specific scores, and EuroQL-5D-5L. RESULTS Our study unveiled a universal and significant expansion and activation of Tregs, without concomitant Teffs activation, across all 13 autoimmune diseases. Both Proleukin® and ready-to-use ILT-101® demonstrated identical effects on Tregs. CGI scores reflecting activity, severity, and efficacy were significantly reduced in the overall patient population. Disease-specific clinical scores improved in five of the six disease cohorts with at least six patients, namely ankylosing spondylitis, systemic lupus erythematosus, Behçet's disease, Sjögren's syndrome, and systemic sclerosis. Urticaria was the only severe adverse event related to treatment. CONCLUSION IL-2LD was well-tolerated, exhibiting specific Treg activation and clinical improvements across the 13 autoimmune diseases. CLINICAL IMPLICATION Tregs stimulation by IL-2LD is a promising therapeutic strategy and IL-2LD holds considerable promise for integration into combinatorial therapeutic approaches.
Collapse
Affiliation(s)
- Roberta Lorenzon
- Assistance Publique - Hopitaux de Paris, Biotherapies Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3) Laboratory, Paris, Île-de-France, France
| | - Claire Ribet
- Assistance Publique - Hopitaux de Paris, Biotherapies Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3) Laboratory, Paris, Île-de-France, France
| | - Fabien Pitoiset
- Assistance Publique - Hopitaux de Paris, Biotherapies Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3) Laboratory, Paris, Île-de-France, France
| | - Selim Aractingi
- Assistance Publique - Hopitaux de Paris, Dermatology Department, Cochin Hospital, Paris, Île-de-France, France
| | - Beatrice Banneville
- Assistance Publique - Hopitaux de Paris, Rheumatology Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France
| | - Laurent Beaugerie
- Assistance Publique - Hopitaux de Paris, Gastroenterology Department, Saint-Antoine Hospital, Paris, Île-de-France, France; Sorbonne Universite, GRC-UPMC 03, Paris, Île-de-France, France
| | - Francis Berenbaum
- Assistance Publique - Hopitaux de Paris, Rheumatology Department, Saint-Antoine Hospital Paris, Île-de-France, France; Sorbonne Universite, INSERM UMR_S938, Paris, Île-de-France, France
| | - Patrice Cacoub
- Assistance Publique - Hopitaux de Paris, Internal Medicine and Clinical Immunology Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France
| | - Julien Champey
- Assistance Publique - Hopitaux de Paris, Rheumatology Department, Saint-Antoine Hospital Paris, Île-de-France, France
| | - Olivier Chazouilleres
- Assistance Publique - Hopitaux de Paris, Hepatology Department, Saint-Antoine Hospital, Paris, Île-de-France, France
| | - Christophe Corpechot
- Assistance Publique - Hopitaux de Paris, Hepatology Department, Saint-Antoine Hospital, Paris, Île-de-France, France
| | - Bruno Fautrel
- Assistance Publique - Hopitaux de Paris, Rheumatology Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, GRC08 - IPLESP, Paris, Île-de-France, France
| | - Arsène Mekinian
- Assistance Publique - Hopitaux de Paris, Internal Medicine Department, Saint-Antoine Hospital, Paris, Île-de-France, France
| | - Elodie Regnier
- Assistance Publique - Hopitaux de Paris, Dermatology Department, Cochin Hospital, Paris, Île-de-France, France
| | - David Saadoun
- Assistance Publique - Hopitaux de Paris, Internal Medicine and Clinical Immunology Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France
| | - Joe-Elie Salem
- INSERM, CIC-1901, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Assistance Publique - Hopitaux de Paris, Pharmacology Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France
| | - Jérémie Sellam
- Assistance Publique - Hopitaux de Paris, Rheumatology Department, Saint-Antoine Hospital Paris, Île-de-France, France; Sorbonne Universite, INSERM UMR_S938, Paris, Île-de-France, France
| | - Philippe Seksik
- Assistance Publique - Hopitaux de Paris, Gastroenterology Department, Saint-Antoine Hospital, Paris, Île-de-France, France; Sorbonne Universite, GRC-UPMC 03, Paris, Île-de-France, France
| | - Eric Vicaut
- Assistance Publique - Hopitaux de Paris, Clinical Research Unit, Lariboisiere Fernand-Widal Hospital, Paris, Île-de-France, France
| | - Michelle Rosenzwajg
- Assistance Publique - Hopitaux de Paris, Biotherapies Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3) Laboratory, Paris, Île-de-France, France
| | - David Klatzmann
- Assistance Publique - Hopitaux de Paris, Biotherapies Department, Pitié-Salpêtrière Hospital, Paris, Île-de-France, France; Sorbonne Universite, INSERM, UMR_S 959, Immunology-Immunopathology- Immunotherapy (i3) Laboratory, Paris, Île-de-France, France.
| |
Collapse
|
7
|
Hoseinzadeh A, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Tavakol Afshari J, Hosseini S, Esmaeili SA. A new generation of mesenchymal stromal/stem cells differentially trained by immunoregulatory probiotics in a lupus microenvironment. Stem Cell Res Ther 2023; 14:358. [PMID: 38072921 PMCID: PMC10712058 DOI: 10.1186/s13287-023-03578-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that multipotent mesenchymal stem/stromal cells (MSCs) are a promising intervention strategy in treating autoimmune inflammatory diseases. It should be stated that systemic immunoregulation is increasingly recognized among the beneficial effects of MSCs and probiotics in treating morbid autoimmune disorders such as lupus. This study aimed to determine if immunoregulatory probiotics L. rhamnosus or L. delbrueckii can change the immunomodulatory effects of MSCs in lupus-like disease. METHODS Pristane-induced lupus (PIL) mice model was created via intraperitoneal injection of Pristane and then confirmed. Naïve MSCs (N-MSCs) were coincubated with two Lactobacillus strains, rhamnosus (R-MSCs) or delbrueckii (D-MSCs), and/or a combination of both (DR-MSCs) for 48 h, then administrated intravenously in separate groups. Negative (PBS-treated normal mice) and positive control groups (PBS-treated lupus mice) were also investigated. At the end of the study, flow cytometry and enzyme-linked immunosorbent assay (ELISA) analysis were used to determine the percentage of Th cell subpopulations in splenocytes and the level of their master cytokines in sera, respectively. Moreover, lupus nephritis was investigated and compared. Analysis of variance (ANOVA) was used for multiple comparisons. RESULTS Abnormalities in serum levels of anti-dsDNA antibodies, creatinine, and urine proteinuria were significantly suppressed by MSCs transplantation, whereas engrafted MSCs coincubation with both L. strains did a lesser effect on anti-dsDNA antibodies. L. rhamnosus significantly escalated the ability of MSCs to scale down the inflammatory cytokines (IFN-ɣ, IL-17), while L. delbrueckii significantly elevated the capacity of MSCs to scale down the percentage of Th cell subpopulations. However, incubation with both strains induced MSCs with augmented capacity in introducing inflammatory cytokines (IFN-ɣ, IL-17). Strikingly, R-MSCs directly restored the serum level of TGF-β more effectively and showed more significant improvement in disease parameters than N-MSCs. These results suggest that R-MSCs significantly attenuate lupus disease by further skew the immune phenotype of MSCs toward increased immunoregulation. CONCLUSIONS Results demonstrated that Lactobacillus strains showed different capabilities in training/inducing new abilities in MSCs, in such a way that pretreated MSCs with L. rhamnosus might benefit the treatment of lupus-like symptoms, given their desirable properties.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Centre, Division of Inflammation and Inflammatory Diseases, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Hosseini
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Zickert A, Janković MR, Malmström V, Chemin K, Gunnarsson I. Occurrence and localization of FOXP3 + cells in kidney biopsies in lupus nephritis and ANCA-associated vasculitis. Clin Rheumatol 2023; 42:2889-2895. [PMID: 37368057 PMCID: PMC10497686 DOI: 10.1007/s10067-023-06676-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
The study aims to increase the understanding regarding the role of regulatory T cells (Tregs) in lupus nephritis (LN) and ANCA-associated vasculitis (AAV) by comparing their localization in renal tissue and changes following immunosuppressive therapy. Kidney biopsies from 12 patients with LN and 7 patients with AAV were examined. Kidney biopsies had been performed both at active disease and following immunosuppressive treatment. Clinical data was collected at both biopsy occasions. Expression of Forkhead Box P 3 (Foxp3) in renal tissue was assessed by immunohistochemistry. An arbitrary scale was used to estimate the number of Foxp3+ cells. In LN, 8/12 (67%) had positive tissue staining for Foxp3 at baseline, most pronounced in inflammatory infiltrates, but also interstitially and in a peri-glomerular pattern. At second biopsies, after immunosuppressive treatment, 4/12 (33%) still had detectable Foxp3+ cells, found in persisting inflammatory infiltrates and some in the interstitium. Patients with a good clinical response to treatment had high grade of Foxp3+ cells in first biopsies. In AAV, only 2/7 (29%) had positive staining for Foxp3 at baseline, in inflammatory infiltrates and to a lesser extent in the interstitium, despite large areas of inflammatory infiltrates in all patients. At follow-up, 2/7 (29%) biopsies were positive for Foxp3. Our data show a higher presence of Foxp3+ cells in renal tissue from LN patients compared to AAV, suggesting that Tregs may be differently involved in the control of inflammatory mechanisms in these diseases. These findings could have further implication for therapeutic approaches aiming at restoring the immunological tolerance. Key Points • Foxp3+-cells are present in larger amount in renal tissue in lupus nephritis vs. ANCA-associated vasculitis. • Our data suggest that Foxp3+ regulatory T cells are involved in the control of inflammatory processes in lupus nephritis.
Collapse
Affiliation(s)
- Agneta Zickert
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden.
- Rheumatology Unit, S-171 76, Karolinska University Hospital, Stockholm, Sweden.
| | - Marija Ratković Janković
- Department of Nephrology, Clinic of Pediatrics, University Clinical Center Nis, Medical Faculty, University of Nis, Nis, Serbia
| | - Vivianne Malmström
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
| | - Karine Chemin
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
| | - Iva Gunnarsson
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Solna, Sweden
- Rheumatology Unit, S-171 76, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Leonard WJ, Lin JX. Strategies to therapeutically modulate cytokine action. Nat Rev Drug Discov 2023; 22:827-854. [PMID: 37542128 DOI: 10.1038/s41573-023-00746-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 08/06/2023]
Abstract
Cytokines are secreted or membrane-presented molecules that mediate broad cellular functions, including development, differentiation, growth and survival. Accordingly, the regulation of cytokine activity is extraordinarily important both physiologically and pathologically. Cytokine and/or cytokine receptor engineering is being widely investigated to safely and effectively modulate cytokine activity for therapeutic benefit. IL-2 in particular has been extensively engineered, to create IL-2 variants that differentially exhibit activities on regulatory T cells to potentially treat autoimmune disease versus effector T cells to augment antitumour effects. Additionally, engineering approaches are being applied to many other cytokines such as IL-10, interferons and IL-1 family cytokines, given their immunosuppressive and/or antiviral and anticancer effects. In modulating the actions of cytokines, the strategies used have been broad, including altering affinities of cytokines for their receptors, prolonging cytokine half-lives in vivo and fine-tuning cytokine actions. The field is rapidly expanding, with extensive efforts to create improved therapeutics for a range of diseases.
Collapse
Affiliation(s)
- Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Tsai YG, Liao PF, Hsiao KH, Wu HM, Lin CY, Yang KD. Pathogenesis and novel therapeutics of regulatory T cell subsets and interleukin-2 therapy in systemic lupus erythematosus. Front Immunol 2023; 14:1230264. [PMID: 37771588 PMCID: PMC10522836 DOI: 10.3389/fimmu.2023.1230264] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous multisystem inflammatory disease with wide variability in clinical manifestations. Natural arising CD4+ regulatory T cells (Tregs) play a critical role in maintaining peripheral tolerance by suppressing inflammation and preventing autoimmune responses in SLE. Additionally, CD8+ regulatory T cells, type 1 regulatory T cells (Tr1), and B regulatory cells also have a less well-defined role in the pathogenesis of SLE. Elucidation of the roles of various Treg subsets dedicated to immune homeostasis will provide a novel therapeutic approach that governs immune tolerance for the remission of active lupus. Diminished interleukin (IL)-2 production is associated with a depleted Treg cell population, and its reversibility by IL-2 therapy provides important reasons for the treatment of lupus. This review focuses on the pathogenesis and new therapeutics of human Treg subsets and low-dose IL-2 therapy in clinical benefits with SLE.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- Department of Pediatrics, Changhua Christian Children’s Hospital, Changhua, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Pei-Fen Liao
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kai-Hung Hsiao
- Department of Allergy, Immunology and Rheumatology, Changhua Christian Hospital, Changhua, Taiwan
| | - Hung-Ming Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ching-Yuang Lin
- Division of Pediatric Nephrology, Children’s Hospital, China Medical University Hospital, Taichung, Taiwan
| | - Kuender D. Yang
- Department of Pediatrics, Mackay Memorial Hospital, New Taipei City, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
11
|
Zhang X, Zhang J, Pan Z, Zhang Y, Xu X, Sheng Y, Zhu Z, Zhou F, Wen L. Transcriptome sequencing reveals novel molecular features of SLE severity. Front Genet 2023; 14:1121359. [PMID: 37554401 PMCID: PMC10406386 DOI: 10.3389/fgene.2023.1121359] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 07/03/2023] [Indexed: 08/10/2023] Open
Abstract
Introduction: Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by the production of autoantibodies, immune complex deposition, and tissue/organ damage. In this study, we aimed to identify molecular features and signaling pathways associated with SLE severity using RNA sequencing (RNA-seq), single-cell RNA sequencing (scRNA-seq), and clinical parameters. Methods: We analyzed transcriptome profiles of 45 SLE patients, grouped into mild (mSLE, SLEDAI ≤ 9) and severe (sSLE, SLEDAI > 9) based on SLE Disease Activity Index (SLEDAI) scores. We also collected clinical data on anti-dsDNA, ANA, ESR, CRP, snRNP, AHA, and anti-Smith antibody status for each patient. Results: By comparing gene expression across groups, we identified 12 differentially expressed genes (DEGs), including 7 upregulated (CEACAM6, UCHL1, ARFGEF3, AMPH, SERPINB10, TACSTD2, and OTX1) and 5 downregulated (SORBS2, TRIM64B, SORCS3, DRAXIN, and PCDHGA10) DEGs in sSLE compared to mSLE. Furthermore, using the CIBERSORT algorithm, we found that Treg cells were significantly decreased in sSLE and negatively correlated with AMPH expression, which was mainly expressed in Treg cells from SLE patients according to public scRNA-seq data (GSE135779). Discussion: Overall, our findings shed light on the molecular mechanisms underlying SLE severity and provide insight into potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Jiali Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Zhaobing Pan
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Yuxi Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Xiaoqing Xu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Yujun Sheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Zhengwei Zhu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Leilei Wen
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
12
|
Zhou X, Qi H, Li M, Li Y, Zhu X, Amin S, Alexander M, Diadhiou C, Davidson A, Zeng H. mTORC2 contributes to systemic autoimmunity. Immunology 2023; 168:554-568. [PMID: 36273262 PMCID: PMC9975033 DOI: 10.1111/imm.13594] [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: 05/27/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
The development of many systemic autoimmune diseases, including systemic lupus erythematosus, is associated with overactivation of the type I interferon (IFN) pathway, lymphopenia and increased follicular helper T (Tfh)-cell differentiation. However, the cellular and molecular mechanisms underlying these immunological perturbations remain incompletely understood. Here, we show that the mechanistic target of rapamycin complex 2 (mTORC2) promotes Tfh differentiation and disrupts Treg homeostasis. Inactivation of mTORC2 in total T cells, but not in Tregs, greatly ameliorated the immunopathology in a systemic autoimmunity mouse model. This was associated with reduced Tfh differentiation, B-cell activation, and reduced T-cell glucose metabolism. Finally, we show that type I IFN can synergize with TCR ligation to activate mTORC2 in T cells, which partially contributes to T-cell lymphopenia. These data indicate that mTORC2 may act as downstream of type I IFN, TCR and costimulatory receptor ICOS, to promote glucose metabolism, Tfh differentiation, and T-cell lymphopenia, but not to suppress Treg function in systemic autoimmunity. Our results suggest that mTORC2 might be a rational target for systemic autoimmunity treatment.
Collapse
Affiliation(s)
- Xian Zhou
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Haiyu Qi
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Rheumatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Meilu Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Dermatology, the Second Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150001, P. R. China
| | - Yanfeng Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Xingxing Zhu
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Shreyasee Amin
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Mariam Alexander
- Division of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN 55905, USA
| | - Catherine Diadhiou
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Hu Zeng
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic Rochester, MN 55905, USA
| |
Collapse
|
13
|
Goldschen L, Ellrodt J, Amonoo HL, Feldman CH, Case SM, Koenen KC, Kubzansky LD, Costenbader KH. The link between post-traumatic stress disorder and systemic lupus erythematosus. Brain Behav Immun 2023; 108:292-301. [PMID: 36535611 PMCID: PMC10018810 DOI: 10.1016/j.bbi.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 12/03/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous, multisystem autoimmune disorder characterized by unpredictable disease flares. Although the pathogenesis of SLE is complex, an epidemiologic link between posttraumatic stress disorder (PTSD) and the development of SLE has been identified, suggesting that stress-related disorders alter the susceptibility to SLE. Despite the strong epidemiologic evidence connecting PTSD and SLE, gaps remain in our understanding of how the two may be connected. Perturbations in the autonomic nervous system, neuroendocrine system, and at the genomic level may cause and sustain immune dysregulation that could lower the threshold for the development and propagation of SLE. We first describe shared risk factors for SLE and PTSD. We then describe potential biological pathways which may facilitate excessive inflammation in the context of PTSD. Among those genetically predisposed to SLE, systemic inflammation that accompanies chronic stress may fan the flames of smoldering SLE by priming immune pathways. Further studies on the connection between trauma and inflammation will provide important data on pathogenesis, risk factors, and novel treatments for SLE.
Collapse
Affiliation(s)
- Lauren Goldschen
- Department of Psychiatry, Brigham and Women's Hospital, 60 Fenwood Road, MA 02115, USA.
| | - Jack Ellrodt
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Hermioni L Amonoo
- Department of Psychiatry, Brigham and Women's Hospital, 60 Fenwood Road, MA 02115, USA; Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Candace H Feldman
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Siobhan M Case
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| |
Collapse
|
14
|
Goswami TK, Singh M, Dhawan M, Mitra S, Emran TB, Rabaan AA, Mutair AA, Alawi ZA, Alhumaid S, Dhama K. Regulatory T cells (Tregs) and their therapeutic potential against autoimmune disorders - Advances and challenges. Hum Vaccin Immunother 2022; 18:2035117. [PMID: 35240914 PMCID: PMC9009914 DOI: 10.1080/21645515.2022.2035117] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/10/2022] [Accepted: 01/22/2022] [Indexed: 02/06/2023] Open
Abstract
Autoimmune diseases are caused when immune cells act against self-protein. This biological self-non-self-discrimination phenomenon is controlled by a distinct group of lymphocytes known as regulatory T cells (Tregs), which are key inflammatory response regulators and play a pivotal role in immune tolerance and homeostasis. Treg-mediated robust immunosuppression provides self-tolerance and protection against autoimmune diseases. However, once this system fails to operate or poorly operate, it leads to an extreme situation where immune system reacts against self-antigens and destroys host organs, thus causing autoimmune diseases. Tregs can target both innate and adaptive immunity via modulating multiple immune cells such as neutrophils, monocytes, antigen-presenting cells, B cells, and T cells. This review highlights the Treg-mediated immunosuppression, role of several markers and their interplay during Treg development and differentiation, and advances in therapeutic aspects of Treg cells to reduce severity of autoimmunity-related conditions along with emphasizing limitations and challenges of their usages.
Collapse
Affiliation(s)
- Tapas Kumar Goswami
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Mithilesh Singh
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- The Trafford Group of Colleges, Manchester, UK
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW, Australia
| | - Zainab Al Alawi
- Division of Allergy and Immunology, College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa, Saudi Arabia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
15
|
Humrich JY, Cacoub P, Rosenzwajg M, Pitoiset F, Pham HP, Guidoux J, Leroux D, Vazquez T, Riemekasten G, Smolen JS, Tsokos G, Klatzmann D. Low-dose interleukin-2 therapy in active systemic lupus erythematosus (LUPIL-2): a multicentre, double-blind, randomised and placebo-controlled phase II trial. Ann Rheum Dis 2022; 81:1685-1694. [PMID: 35973803 DOI: 10.1136/ard-2022-222501] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/20/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES A regulatory T cell (Treg) insufficiency due to shortage of interleukin-2 (IL-2) is central to the pathophysiology of systemic lupus erythematosus (SLE). We performed a multicentre, double-blinded, randomised, placebo-controlled phase II proof-of-concept trial to evaluate the efficacy of low-dose IL-2 therapy in patients with SLE having moderate-to-severe disease activity while receiving standard treatment. METHODS We randomly assigned 100 patients in a 1:1 ratio to receive either 1.5 million IU/day of subcutaneous IL-2 (ILT-101) or placebo for 5 days followed by weekly injections for 12 weeks. Clinical efficacy was assessed at week 12 in a predefined hierarchical analysis of (1) the SLE responder index-4 (SRI-4) response as a primary end point, and of (2) relative and (3) absolute changes in the Safety of Estrogens in Lupus Erythematosus National Assessment-Systemic Lupus Erythematosus Disease Activity Index scores as key secondary end points. RESULTS The primary end point was not met in the intention-to-treat population (ILT-101: 68%, placebo: 58%; p=0.3439), due to a 100% SRI-4 response rate in the placebo group from the two sites from Bulgaria. A post hoc per-protocol analysis on a prespecified population that excluded patients from these two sites (n=53) showed a statistically significant difference for the SRI-4 response rate (ILT-101: 83.3%; placebo: 51.7%; p=0.0168), and for the two key secondary end points, accompanied by differences in several secondary exploratory end points. ILT-101 was well tolerated and there was no generation of antidrug antibodies. CONCLUSIONS The post hoc hierarchical analysis of the primary and key secondary end points in a per-protocol population, complemented by the exploratory analyses of multiple other secondary end points, support that low-dose IL-2 is beneficial in active SLE. TRIAL REGISTRATION NUMBER NCT02955615.
Collapse
Affiliation(s)
- Jens Y Humrich
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Patrice Cacoub
- Service de médecine interne et immunologie clinique, Hopital Pitie-Salpetriere, Paris, France
- Sorbonne Université-INSERM UMRS959, Immunology-Immunopathology-Immunotherapy (i3) laboratory, Sorbonne Universite, Paris, France
| | - Michelle Rosenzwajg
- Sorbonne Université-INSERM UMRS959, Immunology-Immunopathology-Immunotherapy (i3) laboratory, Sorbonne Universite, Paris, France
- Biotherapies Department, Pitié-Salpêtrière hospital, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Fabien Pitoiset
- Sorbonne Université-INSERM UMRS959, Immunology-Immunopathology-Immunotherapy (i3) laboratory, Sorbonne Universite, Paris, France
- Biotherapies Department, Pitié-Salpêtrière hospital, Assistance Publique-Hopitaux de Paris, Paris, France
| | | | | | | | | | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | - George Tsokos
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - David Klatzmann
- Sorbonne Université-INSERM UMRS959, Immunology-Immunopathology-Immunotherapy (i3) laboratory, Sorbonne Universite, Paris, France
- Biotherapies Department, Pitié-Salpêtrière hospital, Assistance Publique-Hopitaux de Paris, Paris, France
| |
Collapse
|
16
|
Zhao X, Wang S, Wang S, Xie J, Cui D. mTOR signaling: A pivotal player in Treg cell dysfunction in systemic lupus erythematosus. Clin Immunol 2022; 245:109153. [DOI: 10.1016/j.clim.2022.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
17
|
Robinson GA, Peng J, Peckham H, Butler G, Pineda-Torra I, Ciurtin C, Jury EC. Investigating sex differences in T regulatory cells from cisgender and transgender healthy individuals and patients with autoimmune inflammatory disease: a cross-sectional study. THE LANCET. RHEUMATOLOGY 2022; 4:e710-e724. [PMID: 36353692 PMCID: PMC9633330 DOI: 10.1016/s2665-9913(22)00198-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Sexual dimorphisms, which vary depending on age group and pubertal status, have been described across both the innate and adaptive immune system. We explored the influence of sex hormones on immune phenotype in the context of adolescent health and autoimmunity. Methods In this cross-sectional study, healthy, post-pubertal cisgender individuals (aged 16-25 years); healthy, pre-pubertal cisgender individuals (aged 6-11 years); transgender individuals (aged 18-19 years) undergoing gender-affirming treatment (testosterone in individuals assigned female sex at birth and oestradiol in individuals assigned male sex at birth); and post-pubertal cisgender individuals (aged 14-25 years) with juvenile-onset systemic lupus erythematosus (SLE) age-matched to cisgender individuals without juvenile-onset SLE were eligible for inclusion. Frequencies of 28 immune-cell subsets (including different T cell, B cell, and monocyte subsets) from each participant were measured in peripheral blood mononuclear cells by flow cytometry and analysed by balanced random forest machine learning. RNA-sequencing was used to compare sex and gender differences in regulatory T (Treg) cell phenotype between participants with juvenile-onset SLE, age-matched cis-gender participants without the disease, and age matched transgender individuals on gender-affirming sex hormone treatment. Differentially expressed genes were analysed by cluster and pathway analysis. Suppression assays assessed the anti-inflammatory function of Treg cells in vitro. Findings Between Sept 5, 2012, and Nov 6, 2019, peripheral blood was collected from 39 individuals in the post-pubertal group (17 [44%] cisgender men, mean age 18·76 years [SD 2·66]; 22 [56%] cisgender women, mean age 18·59 years [2·81]), 14 children in the cisgender pre-pubertal group (seven [50%] cisgender boys, mean age 8·90 [1·66]; seven [50%] cisgender girls, mean age 8·40 [1·58]), ten people in the transgender group (five [50%] transgender men, mean age 18·20 years [0·47]; five [50%] transgender women, mean age 18·70 years [0·55]), and 35 people in the juvenile-onset SLE group (12 [34%] cisgender men, mean age 18·58 years [2·35]; 23 [66%] cisgender women, mean age 19·48 [3·08]). Statistically significantly elevated frequencies of Treg cells were one of the top immune-cell features differentiating young post-pubertal cisgender men from similarly aged cisgender women (p=0·0097). Treg cells from young cisgender men had a statistically significantly increased suppressive capacity in vitro compared with those from cisgender women and a distinct transcriptomic signature significantly enriched for genes in the PI3K-AKT signalling pathway. Gender-affirming sex hormones in transgender men and transgender women induced multiple statistically significant changes in the Treg-cell transcriptome, many of which enriched functional pathways that overlapped with those altered between cisgender men and cisgender women, highlighting a hormonal influence on Treg-cell function by gender. Finally, sex differences in Treg-cell frequency were absent and suppressive capacity was reversed in patients with juvenile-onset SLE, but sex differences in Treg-cell transcriptional signatures were significantly more pronounced in patients with juvenile-onset SLE compared with individuals without juvenile-onset SLE, suggesting that sex hormone signalling could be dysregulated in autoimmunity. Interpretation Sex-chromosomes and hormones might drive changes in Treg-cell frequency and function. Young post-pubertal men have a more anti-inflammatory Treg-cell profile, which could explain inflammatory disease susceptibilities, and inform sex-tailored therapeutic strategies. Funding Versus Arthritis, UK National Institute for Health Research University College London Hospital Biomedical Research Centre, Lupus UK, and The Rosetrees Trust.
Collapse
Affiliation(s)
- George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Gary Butler
- Department of Paediatric and Adolescent Endocrinology, University College London Hospital and Great Ormond Street Institute of Child Health, University College London, London, UK
- Gender Identity Development Service, Tavistock and Portman NHS Foundation Trust, London, UK
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| |
Collapse
|
18
|
Li M, Luo L, Wu Y, Song Z, Ni B, Hao F, Luo N. Elevated apoptosis and abnormal apoptosis signaling of regulatory T cells in patients with systemic lupus erythematosus. Lupus 2022; 31:1441-1455. [PMID: 35950636 DOI: 10.1177/09612033221119455] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In systemic lupus erythematosus (SLE), immune tolerance is influenced by defects in naturally occurring T cells (Tregs). To investigate the apoptosis rate of Tregs and their suppressive activity in patients with SLE and then to recognize the genes and signaling pathways that cause Treg apoptosis. FACS was used to assess the frequency and apoptosis rates of Tregs in 48 SLE patients and 28 normal controls (NCs). Coculture of Tregs with CD4+CD25-CD127dim/- T cells was used to assess the suppressive activity of Tregs. Microarray analysis was used to generate unstimulated Tregs gene expression profiles from very high activity patients with SLE and NCs. Real-time PCR was used to confirm differential gene expression. In patients with SLE, the frequency of Tregs was substantially reduced compared to Tregs from NCs. Furthermore, Tregs from SLE patients had an elevated rate of apoptosis and a lower suppressing ability than Tregs from NCs. Tregs apoptosis was negatively associated with the total count of Tregs and positively related to disease activity. Unstimulated Tregs gene expression profiles from patients with recent-onset SLE revealed a biological response that can cause apoptosis, partially triggered by stress, DNA damage, and cytokine stimulation. The discovery of pathway-specific expression signatures is a significant step forward in understanding how Tregs defects contribute to the pathogenesis of SLE. Our findings may contribute to the development of new strategies for treating SLE based on abnormal Tregs apoptosis and restoring immune homeostasis in patients with SLE.
Collapse
Affiliation(s)
- Mingfang Li
- Department of Dermatology, 117980The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Southwest Hospital, PLA, 12525Third Military Medical University, Chongqing, China
| | - Li Luo
- Department of Dermatology, Southwest Hospital, PLA, 12525Third Military Medical University, Chongqing, China
| | - Yi Wu
- Digital Medicine Institute, Biomedical Engineering College, PLA, 12525Third Military Medical University, Chongqing, China
| | - Zhiqiang Song
- Department of Dermatology, Southwest Hospital, PLA, 12525Third Military Medical University, Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude MilitaryMedicine, PLA, 12525Third Military Medical University, Chongqing, China
| | - Fei Hao
- Department of Dermatology, Southwest Hospital, PLA, 12525Third Military Medical University, Chongqing, China
| | - Na Luo
- Department of Dermatology, Southwest Hospital, PLA, 12525Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
Caza T, Wijewardena C, Al-Rabadi L, Perl A. Cell type-specific mechanistic target of rapamycin-dependent distortion of autophagy pathways in lupus nephritis. Transl Res 2022; 245:55-81. [PMID: 35288362 PMCID: PMC9240418 DOI: 10.1016/j.trsl.2022.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 01/02/2023]
Abstract
Pro-inflammatory immune system development, metabolomic defects, and deregulation of autophagy play interconnected roles in driving the pathogenesis of systemic lupus erythematosus (SLE). Lupus nephritis (LN) is a leading cause of morbidity and mortality in SLE. While the causes of SLE have not been clearly delineated, skewing of T and B cell differentiation, activation of antigen-presenting cells, production of antinuclear autoantibodies and pro-inflammatory cytokines are known to contribute to disease development. Underlying this process are defects in autophagy and mitophagy that cause the accumulation of oxidative stress-generating mitochondria which promote necrotic cell death. Autophagy is generally inhibited by the activation of the mammalian target of rapamycin (mTOR), a large protein kinase that underlies abnormal immune cell lineage specification in SLE. Importantly, several autophagy-regulating genes, including ATG5 and ATG7, as well as mitophagy-regulating HRES-1/Rab4A have been linked to lupus susceptibility and molecular pathogenesis. Moreover, genetically-driven mTOR activation has been associated with fulminant lupus nephritis. mTOR activation and diminished autophagy promote the expansion of pro-inflammatory Th17, Tfh and CD3+CD4-CD8- double-negative (DN) T cells at the expense of CD8+ effector memory T cells and CD4+ regulatory T cells (Tregs). mTOR activation and aberrant autophagy also involve renal podocytes, mesangial cells, endothelial cells, and tubular epithelial cells that may compromise end-organ resistance in LN. Activation of mTOR complexes 1 (mTORC1) and 2 (mTORC2) has been identified as biomarkers of disease activation and predictors of disease flares and prognosis in SLE patients with and without LN. This review highlights recent advances in molecular pathogenesis of LN with a focus on immuno-metabolic checkpoints of autophagy and their roles in pathogenesis, prognosis and selection of targets for treatment in SLE.
Collapse
Affiliation(s)
| | - Chathura Wijewardena
- Departments of Medicine, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York
| | - Laith Al-Rabadi
- Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Andras Perl
- Departments of Medicine, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York; Biochemistry and Molecular Biology, Neuroscience and Physiology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York; Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York.
| |
Collapse
|
20
|
Rao J, Li S, Wang Q, Cheng Q, Ji Y, Fu W, Huang H, Shi L, Wu X. Comparison of Peripheral Blood Regulatory T Cells and Functional Subsets Between Ocular and Generalized Myasthenia Gravis. Front Med (Lausanne) 2022; 9:851808. [PMID: 35755064 PMCID: PMC9218215 DOI: 10.3389/fmed.2022.851808] [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: 01/10/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose This study aims to discuss the function mechanism of regulatory T cells and its subsets in the pathogenic process of myasthenia gravis by contracting the activation levels of those cells in peripheral blood among healthy people, patients with ocular myasthenia gravis (oMG) and patients with generalized myasthenia gravis (gMG). Method Healthy people, newly diagnosed oMG patients, and gMG patients were enrolled in this study. The percentage of the CD3+CD4+CD25+ Treg cells, CD3+CD4+CD25+Foxp3+ Treg cells, CD3+CD4+CD25+Foxp3hi CD45RA–aTreg cells, CD3+CD4+CD25+Foxp3loCD45RA–n-sTreg cells, and CD3+CD4+CD25+ Foxp3loCD45RA+rTreg cells in the peripheral blood were examined by flow cytometry. And then analyzed the differences of Treg cells and its subsets among the study members. Results The percentage of the CD4+CD25+Treg cells in the peripheral blood of oMG patients and gMG patients were both lower than that of healthy people (p < 0.05), the percentage of patients with oMG had no distinct difference with that of patients with gMG (p = 0.475), however. Also, the percentage of CD3+CD4+CD25+Foxp3+Treg cells in the oMG and gMG patients’ group were both lower than that of healthy group. And the percentage of CD25+Foxp3+Treg cells in the peripheral blood of patients with oMG and healthy people were both higher than that of patients with gMG (p < 0.05). The percentage of rTreg in the CD3+CD4+CD25+Treg of the peripheral blood for both gMG and oMG patients’ group were lower than healthy group (p < 0.05), but there was no statistical significance between the oMG and gMG patients’ group (p = 0.232). The percentage of the aTreg cells in the CD3+CD4+CD25+Treg cells of the peripheral blood for the oMG patients was higher than that of gMG patients (p < 0.05), but both of them were lower than healthy group (p < 0.05). The percentage of n-sTreg cells in the peripheral blood descended among the gMG patients’ group, oMG patients’ group, and healthy group (p < 0.05). Conclusion The changes in the number and function of Treg cells and its subsets can cause the impairment of negative immune regulation, which may mediate the triggering of oMG and its progression to gMG.
Collapse
Affiliation(s)
- Jie Rao
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Siyu Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiyu Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qi Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenwen Fu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Huang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ling Shi
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaorong Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
21
|
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
|
22
|
Yang Y, Wang C, Shi L, Yang S, Liu Y, Luo J, Wang C. Clinical Characteristics and CD4+ T Cell Subsets in IgG4-Related Disease. Front Immunol 2022; 13:825386. [PMID: 35432312 PMCID: PMC9010737 DOI: 10.3389/fimmu.2022.825386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
Objectives To characterize the clinical features of IgG4-related disease (IgG4-RD) and analyze the peripheral T lymphocyte subsets and cytokine levels. Methods A total of 52 patients with newly diagnosed IgG4-RD were enrolled in the retrospective study. Baseline clinical characteristics and examinational findings were systematically reviewed. Results IgG4-RD patients had a male predominance, with an average age of 57.4 ± 10.3 years (range 27-81). The mean number of involved organs was 2.7 (range 1-8). Submandibular gland (57.7%) and lacrimal gland/orbit (40.4%) were the most commonly involved organs. Serum IgG4 increased in 97.9% of the patients, the median level was 1300 (585.25, 1975) mg/dl. Decreased C3 and C4 accounted for 77.8% and 55.6% of this patient cohort, respectively. Receiver operating characteristic (ROC) test indicated the possibility of lung/pleura involvement when C3 was less than 0.570 g/l (AUC = 0.788, P = 0.014), and kidney involvement when C3 was less than 0.545 g/l (AUC = 0.796, P = 0.014). Compared with healthy controls (HC), the absolute Th1 counts were higher in IgG4-RD patients (157.58 cells/μl vs. 130.54 cells/μl, P = 0.038), while the absolute counts of Th2, Th17 and T regulatory (Treg) cells, as well as Th17/Treg ratio were not statistically different. The levels of serum IL-4, IL-6, IL-10, IL-17, TNF-α, and IFN-γ were higher in patients with IgG4-RD as compared with HC (P < 0.001). Serum IL-10 was positively correlated with IL-4, TNF-α and IFN-γ, but uncorrelated with Treg cells. Serum IgG4 level was positively associated with the number of affected organs, eosinophil counts, and ESR, whereas inversely associated with C3, C4, IgM, and IgA. Conclusion Submandibular and lacrimal glands are the most commonly involved organs in IgG4-RD. Serum C3 level could be a predictor of lung/pleura and kidney involvement in the disease process. Elevated Th1 cells are probably related to chronic inflammation and fibrosis. Treg cells are unlikely to play an important role in the pathogenesis of IgG4-RD.
Collapse
Affiliation(s)
- Yan Yang
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chen Wang
- Department of Pathology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Shi
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuoran Yang
- Department of Oral and Maxillofacial Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Liu
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Luo
- Department of Rheumatology Laboratory, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Caihong Wang
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Caihong Wang,
| |
Collapse
|
23
|
Fanton C, Furie R, Chindalore V, Levin R, Diab I, Dixit N, Haglund C, Gibbons J, Hanan N, Dickerson D, Zalevsky J, Kotzin BL. Selective expansion of regulatory T cells by NKTR-358 in healthy volunteers and patients with systemic lupus erythematosus. J Transl Autoimmun 2022; 5:100152. [PMID: 35517914 PMCID: PMC9062472 DOI: 10.1016/j.jtauto.2022.100152] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 11/28/2022] Open
Abstract
Objective To evaluate NKTR-358, a polyethylene glycol-interleukin-2 conjugate composition designed to selectively induce regulatory T cells (Tregs), in first-in-human studies. Methods Healthy volunteers and patients with systemic lupus erythematosus (SLE) received single- or multiple-dose (biweekly) NKTR-358 or placebo in two sequential, randomized, phase 1 studies (single ascending dose [SAD; NCT04133116] and multiple ascending dose [MAD; NCT03556007]). Primary objectives were safety and tolerability; secondary objectives included pharmacokinetics (PK) and immune effects of NKTR-358; exploratory objectives included effects on SLE disease activity. Results There were eight ascending dose cohorts in the SAD study (0.3-28.0 μg/kg: n = 76; placebo: n = 24) and four in the MAD study (3-24.0 μg/kg: n = 36; placebo: n = 12). Most adverse events (AEs) were grade 1-2 injection-site reactions, with no treatment-related serious or severe AEs, or deaths. PK data showed dose proportionality and prolonged exposure (mean half-life: 7.4-12.9 days). Dose-dependent, selective, and sustained increases in percentages and absolute numbers of total CD4+ Tregs and CD25bright Tregs were observed, with no significant changes in conventional CD4+ and CD8+ T cells, and low-level increases in natural killer cells. At the highest doses tested, administration of NKTR-358 resulted in a 12-17-fold increase in CD25bright Tregs over baseline that was sustained for 20-30 days. Conclusion NKTR-358 was well tolerated, had a suitable PK profile for biweekly dosing, and led to marked and selective dose-dependent increases in CD25bright Tregs, with no significant changes in conventional T cells. These results provide strong support for further testing in SLE and other inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | - Robert Levin
- Clinical Research of West Florida, Clearwater, FL, USA
| | - Isam Diab
- Paramount Medical Research, Middleburg Heights, OH, USA
| | - Neha Dixit
- Nektar Therapeutics, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Lerkvaleekul B, Apiwattanakul N, Tangnararatchakit K, Jirapattananon N, Srisala S, Vilaiyuk S. Associations of lymphocyte subpopulations with clinical phenotypes and long-term outcomes in juvenile-onset systemic lupus erythematosus. PLoS One 2022; 17:e0263536. [PMID: 35130317 PMCID: PMC8820627 DOI: 10.1371/journal.pone.0263536] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Juvenile-onset systemic lupus erythematosus (JSLE) is a complex and heterogeneous immune-mediated disease. Cellular components have crucial roles in disease phenotypes and outcomes. We aimed to determine the associations of lymphocyte subsets with clinical manifestations and long-term outcomes in JSLE patients. METHODS A cohort of 60 JSLE patients provided blood samples during active disease, of whom 34 provided further samples during inactive disease. In a longitudinal study, blood samples were obtained from 49 of the JSLE patients at 0, 3, and 6 months. The healthy control (HC) group consisted of 42 age-matched children. Lymphocyte subsets were analyzed by flow cytometry. RESULTS The percentages of CD4+ T, γδ T, and NK cells were significantly decreased in JSLE patients compared with HC, while the percentages of CD8+ T, NKT, and CD19+ B cells were significantly increased. The percentage of regulatory T cells (Tregs) was significantly lower in JSLE patients with lupus nephritis (LN) than in non-LN JSLE patients and HC. The patients were stratified into high and low groups by the median frequency of each lymphocyte subset. The γδ T cells high group and NK cells high group were significantly related to mucosal ulcer. The CD4+ T cells high group was significantly associated with arthritis, and the NKT cells high group was substantially linked with autoimmune hemolytic anemia. The CD8+ T cells low group was mainly related to vasculitis, and the Tregs low group was significantly associated with LN. The percentage of Tregs was significantly increased at 6 months of follow-up, and the LN JSLE group had a lower Treg percentage than the non-LN JSLE group. Predictors of remission on therapy were high Tregs, high absolute lymphocyte count, direct Coombs test positivity, and LN absence at enrollment. CONCLUSION JSLE patients exhibited altered lymphocyte subsets, which were strongly associated with clinical phenotypes and long-term outcomes.
Collapse
Affiliation(s)
- Butsabong Lerkvaleekul
- Faculty of Medicine Ramathibodi Hospital, Division of Rheumatology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Nopporn Apiwattanakul
- Faculty of Medicine Ramathibodi Hospital, Division of Infectious Disease, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Kanchana Tangnararatchakit
- Faculty of Medicine Ramathibodi Hospital, Division of Nephrology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Nisa Jirapattananon
- Faculty of Medicine Ramathibodi Hospital, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Supanart Srisala
- Faculty of Medicine Ramathibodi Hospital, Research Center, Mahidol University, Bangkok, Thailand
| | - Soamarat Vilaiyuk
- Faculty of Medicine Ramathibodi Hospital, Division of Rheumatology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
25
|
Robinson GA, Wilkinson MGL, Wincup C. The Role of Immunometabolism in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2022; 12:806560. [PMID: 35154082 PMCID: PMC8826250 DOI: 10.3389/fimmu.2021.806560] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder in which pathogenic abnormalities within both the innate and adaptive immune response have been described. In order to activated, proliferate and maintain this immunological response a drastic upregulation in energy metabolism is required. Recently, a greater understanding of these changes in cellular bioenergetics have provided new insight into the links between immune response and the pathogenesis of a number of diseases, ranging from cancer to diabetes and multiple sclerosis. In this review, we highlight the latest understanding of the role of immunometabolism in SLE with particular focus on the role of abnormal mitochondrial function, lipid metabolism, and mTOR signaling in the immunological phenomenon observed in the SLE. We also consider what implications this has for future therapeutic options in the management of the disease in future.
Collapse
Affiliation(s)
- George Anthony Robinson
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom.,Department of Rheumatology, University College London Great Ormond Street Institute of Child Health, Infection, Immunity and Inflammation Research and Teaching Department, University College London, London, United Kingdom
| | - Chris Wincup
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| |
Collapse
|
26
|
HSPB5 suppresses renal inflammation and protects lupus-prone NZB/W F1 mice from severe renal damage. Arthritis Res Ther 2022; 24:267. [PMID: 36510250 PMCID: PMC9743758 DOI: 10.1186/s13075-022-02958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Lupus nephritis (LN) is an inflammatory disease of the kidneys affecting patients with systemic lupus erythematosus. Current immunosuppressive and cytotoxic therapies are associated with serious side effects and fail to protect 20-40% of LN patients from end-stage renal disease. In this study, we investigated whether a small heat shock protein, HSPB5, can reduce kidney inflammation and the clinical manifestations of the disease in NZB/W F1 mice. Furthermore, we investigated whether HSPB5 can enhance the effects of methylprednisolone, a standard-of-care drug in LN, in an endotoxemia mouse model. METHODS NZB/W F1 mice were treated with HSPB5, methylprednisolone, or vehicle from 23 to 38 weeks of age. Disease progression was evaluated by weekly proteinuria scores. At the end of the study, the blood, urine, spleens, and kidneys were collected for the assessment of proteinuria, blood urea nitrogen, kidney histology, serum IL-6 and anti-dsDNA levels, immune cell populations, and their phenotypes, as well as the transcript levels of proinflammatory chemokine/cytokines in the kidneys. HSPB5 was also evaluated in combination with methylprednisolone in a lipopolysaccharide-induced endotoxemia mouse model; serum IL-6 levels were measured at 24 h post-endotoxemia induction. RESULTS HSPB5 significantly reduced terminal proteinuria and BUN and substantially improved kidney pathology. Similar trends, although to a lower extent, were observed with methylprednisolone treatment. Serum IL-6 levels and kidney expression of BAFF, IL-6, IFNγ, MCP-1 (CCL2), and KIM-1 were reduced, whereas nephrin expression was significantly preserved compared to vehicle-treated mice. Lastly, splenic Tregs and Bregs were significantly induced with HSPB5 treatment. HSPB5 in combination with methylprednisolone also significantly reduced serum IL-6 levels in endotoxemia mice. CONCLUSIONS HSPB5 treatment reduces kidney inflammation and injury, providing therapeutic benefits in NZB/W F1 mice. Given that HSPB5 enhances the anti-inflammatory effects of methylprednisolone, there is a strong interest to develop HSBP5 as a therapeutic for the treatment of LN.
Collapse
|
27
|
田 佳, 张 霞, 程 功, 刘 庆, 王 世, 何 菁. [Serum interleukin-2 receptor α as a clinical biomarker in patients with systemic lupus erythematosus]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2021; 53:1083-1087. [PMID: 34916686 PMCID: PMC8695147 DOI: 10.19723/j.issn.1671-167x.2021.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To investigate the clinical relevance of serum interleukin-2 receptor α (IL-2Rα) in patients with systemic lupus erythematosus (SLE). METHODS One hundred and seven SLE patients and 39 healthy controls with comparable age and gender were recruited at Peking University People's Hospital from January 2019 to December 2020. Complete clinical data in 107 SLE patients at baseline and follow-up were collected. SLE disease activity index 2000 (SLEDAI-2K) was used to assess the disease activity of the SLE patients. The serum level of IL-2Rα in the SLE patients and healthy controls was measured using enzyme-linked immunosorbent assay (ELISA). The association between serum IL-2Rα and clinical and laboratory parameters was investigated. Mann-Whitney U test or t test, Chi-square test and Spearman correlation were used for statistical analysis. RESULTS The serum IL-2Rα levels were significantly higher in the SLE patients [830.82 (104.2-8 940.48) ng/L], compared with those in the healthy controls [505.1 (78.65-1 711.52) ng/L] (P < 0.001). Association analysis showed that the increased serum IL-2Rα was positively associated with SLEDAI-2K scores and anti-nucleosome antibody (r=0.357, P < 0.001; r=0.25, P=0.027, respectively). Thirty-six of 107 (33.6%) SLE patients had lupus nephritis. Serum IL-2Rα levels were significantly higher in the patients accompanied with lupus nephritis [1 102.14 (126.52-8 940.48) ng/L] than in the patients without lupus nephritis [743.89 (104.19-4 872.06) ng/L] (P=0.032). The patients in the high IL-2Rα group had more lupus nephritis compared with those in the low IL-2Rα group (40.8% vs. 19.4%, P=0.031). Meanwhile, SLEDAI-2K scores were found significantly higher in the high IL-2Rα group than in the low IL-2Rα group [10 (3-21) vs. 7 (3-16), P=0.001]. With the improvement of disease activity in the SLE patients after conventional treatments, serum levels of IL-2Rα [1 119.1 (372.25-2 608.86) ng/L] in the week 12 decreased significantly compared with the baseline [1 556.73 (373.08-8 940.48) ng/L] (P=0.042). CONCLUSION Serum IL-2Rα may be used as a biomarker of disease activity in patients with SLE. There is certain correlation between serum IL-2Rα and renal involvement in SLE.
Collapse
Affiliation(s)
- 佳宜 田
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 霞 张
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 功 程
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 庆红 刘
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 世阳 王
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - 菁 何
- />北京大学人民医院风湿免疫科,北京 100044Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
28
|
Abstract
Failure of regulatory T (Treg) cells to properly control immune responses leads invariably to autoimmunity and organ damage. Decreased numbers or impaired function of Treg cells, especially in the context of inflammation, has been documented in many human autoimmune diseases. Restoration of Treg cell fitness and/or expansion of their numbers using low-dose natural IL-2, the main cytokine driving Treg cell survival and function, has demonstrated clinical efficacy in early clinical trials. Genetically modified IL-2 with an extended half-life and increased selectivity for Treg cells is now in clinical development. Administration of IL-2 combined with therapies targeting other pathways involved in the expression of autoimmune diseases should further enhance its therapeutic potential. Ongoing clinical efforts that capitalize on the early clinical success of IL-2 treatment should bring the use of this cytokine to the forefront of biological treatments for autoimmune diseases.
Collapse
|
29
|
Singh RP, Bischoff DS, Hahn BH. CD8 + T regulatory cells in lupus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:147-156. [PMID: 35880241 PMCID: PMC9242525 DOI: 10.2478/rir-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 04/11/2023]
Abstract
T regulatory cells (Tregs) have a key role in the maintenance of immune homeostasis and the regulation of immune tolerance by preventing the inflammation and suppressing the autoimmune responses. Numerical and functional deficits of these cells have been reported in systemic lupus erythematosus (SLE) patients and mouse models of SLE, where their imbalance and dysregulated activities have been reported to significantly influence the disease pathogenesis, progression and outcomes. Most studies in SLE have focused on CD4+ Tregs and it has become clear that a critical role in the control of immune tolerance after the breakdown of self-tolerance is provided by CD8+ Tregs. Here we review the role, cellular and molecular phenotypes, and mechanisms of action of CD8+ Tregs in SLE, including ways to induce these cells for immunotherapeutic modulation in SLE.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bevra H. Hahn
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
30
|
Li W, Gong M, Park YP, Elshikha AS, Choi SC, Brown J, Kanda N, Yeh WI, Peters L, Titov AA, Teng X, Brusko TM, Morel L. Lupus susceptibility gene Esrrg modulates regulatory T cells through mitochondrial metabolism. JCI Insight 2021; 6:e143540. [PMID: 34156979 PMCID: PMC8410062 DOI: 10.1172/jci.insight.143540] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 06/16/2021] [Indexed: 01/31/2023] Open
Abstract
Estrogen-related receptor γ (Esrrg) is a murine lupus susceptibility gene associated with T cell activation. Here, we report that Esrrg controls Tregs through mitochondria homeostasis. Esrrg deficiency impaired the maintenance and function of Tregs, leading to global T cell activation and autoimmunity in aged mice. Further, Esrrg-deficient Tregs presented an impaired differentiation into follicular Tregs that enhanced follicular helper T cells' responses. Mechanistically, Esrrg-deficient Tregs presented with dysregulated mitochondria with decreased oxygen consumption as well as ATP and NAD+ production. In addition, Esrrg-deficient Tregs exhibited decreased phosphatidylinositol and TGF-β signaling pathways and increased mTOR complex 1 activation. We found that the expression of human ESRRG, which is high in Tregs, was lower in CD4+ T cells from patients with lupus than in healthy controls. Finally, knocking down ESRRG in Jurkat T cells decreased their metabolism. Together, our results reveal a critical role of Esrrg in the maintenance and metabolism of Tregs, which may provide a genetic link between lupus pathogenesis and mitochondrial dysfunction in T cells.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Minghao Gong
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Yuk Pheel Park
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ahmed S Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Egypt
| | - Seung-Chul Choi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Josephine Brown
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Nathalie Kanda
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Wen-I Yeh
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Leeana Peters
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Anton A Titov
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Xiangyu Teng
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
31
|
Katsuyama T, Moulton VR. Splicing factor SRSF1 is indispensable for regulatory T cell homeostasis and function. Cell Rep 2021; 36:109339. [PMID: 34233194 PMCID: PMC8476088 DOI: 10.1016/j.celrep.2021.109339] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 02/24/2021] [Accepted: 06/14/2021] [Indexed: 02/02/2023] Open
Abstract
The ability of regulatory T (Treg) cells to control the immune response and limit the development of autoimmune diseases is determined by distinct molecular processes, which are not fully understood. We show here that serine/arginine-rich splicing factor 1 (SRSF1), which is decreased in T cells from patients with systemic lupus erythematosus, is necessary for the homeostasis and proper function of Treg cells, because its conditional absence in these cells leads to profound autoimmunity and organ inflammation by elevating the glycolytic metabolism and mTORC1 activity and the production of proinflammatory cytokines. Our data reveal a molecular mechanism that controls Treg cell plasticity and offer insights into the pathogenesis of autoimmune disease.
Collapse
Affiliation(s)
- Takayuki Katsuyama
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Graßhoff H, Comdühr S, Monne LR, Müller A, Lamprecht P, Riemekasten G, Humrich JY. Low-Dose IL-2 Therapy in Autoimmune and Rheumatic Diseases. Front Immunol 2021; 12:648408. [PMID: 33868284 PMCID: PMC8047324 DOI: 10.3389/fimmu.2021.648408] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Treg) are crucial for the maintenance of peripheral tolerance and for the control of ongoing inflammation and autoimmunity. The cytokine interleukin-2 (IL-2) is essentially required for the growth and survival of Treg in the peripheral lymphatic tissues and thus plays a vital role in the biology of Treg. Most autoimmune and rheumatic diseases exhibit disturbances in Treg biology either at a numerical or functional level resulting in an imbalance between protective and pathogenic immune cells. In addition, in some autoimmune diseases, a relative deficiency of IL-2 develops during disease pathogenesis leading to a disturbance of Treg homeostasis, which further amplifies the vicious cycle of tolerance breach and chronic inflammation. Low-dose IL-2 therapy aims either to compensate for this IL-2 deficiency to restore a physiological state or to strengthen the Treg population in order to be more effective in counter-regulating inflammation while avoiding global immunosuppression. Here we highlight key findings and summarize recent advances in the clinical translation of low-dose IL-2 therapy for the treatment of autoimmune and rheumatic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jens Y. Humrich
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany
| |
Collapse
|
33
|
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
|
34
|
Takasato Y, Kurashima Y, Kiuchi M, Hirahara K, Murasaki S, Arai F, Izawa K, Kaitani A, Shimada K, Saito Y, Toyoshima S, Nakamura M, Fujisawa K, Okayama Y, Kunisawa J, Kubo M, Takemura N, Uematsu S, Akira S, Kitaura J, Takahashi T, Nakayama T, Kiyono H. Orally desensitized mast cells form a regulatory network with Treg cells for the control of food allergy. Mucosal Immunol 2021; 14:640-651. [PMID: 33299086 PMCID: PMC8075951 DOI: 10.1038/s41385-020-00358-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 10/09/2020] [Accepted: 10/21/2020] [Indexed: 02/04/2023]
Abstract
Oral immunotherapy (OIT) is an effective approach to controlling food allergy. Although the detailed molecular and cellular mechanisms of OIT are unknown currently, they must be understood to advance the treatment of allergic diseases in general. To elucidate the mechanisms of OIT, especially during the immunological transition from desensitization to allergy regulation, we generated a clinical OIT murine model and used it to examine immunological events of OIT. We found that in mice that completed OIT successfully, desensitized mast cells (MCs) showed functionally beneficial alterations, such as increased induction of regulatory cytokines and enhanced expansion of regulatory T cells. Importantly, these regulatory-T-cell-mediated inhibitions of allergic responses were dramatically decreased in mice lacking OIT-induced desensitized MC. Collectively, these findings show that the desensitization process modulates the activation of MCs, leading directly to enhanced induction of regulatory-T-cell expansion and promotion of clinical allergic unresponsiveness. Our results suggest that efficiently inducing regulatory MCs is a novel strategy for the treatment of allergic disease.
Collapse
Affiliation(s)
- Yoshihiro Takasato
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.26091.3c0000 0004 1936 9959Department of Pediatrics, Keio University School of Medicine, Tokyo, 160-8582 Japan
| | - Yosuke Kurashima
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.136304.30000 0004 0370 1101Department of Innovative Medicine and Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan ,grid.266100.30000 0001 2107 4242Division of Gastroenterology, Department of Medicine, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA 92093-0956 USA ,grid.136304.30000 0004 0370 1101Institute for Global Prominent Research, Chiba University, Chiba, 260-8670 Japan ,grid.482562.fLaboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085 Japan
| | - Masahiro Kiuchi
- grid.136304.30000 0004 0370 1101Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan
| | - Kiyoshi Hirahara
- grid.136304.30000 0004 0370 1101Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan
| | - Sayuri Murasaki
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan
| | - Fujimi Arai
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan
| | - Kumi Izawa
- grid.258269.20000 0004 1762 2738Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421 Japan
| | - Ayako Kaitani
- grid.258269.20000 0004 1762 2738Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421 Japan
| | - Kaoru Shimada
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan
| | - Yukari Saito
- grid.136304.30000 0004 0370 1101Department of Innovative Medicine and Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan
| | - Shota Toyoshima
- grid.260969.20000 0001 2149 8846Allergy and Immunology Research Project Team, Research Institute of Medical Science, Center for Allergy, Center for Medical Education, Nihon University School of Medicine, Tokyo, 173-8610 Japan
| | - Miho Nakamura
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Kumiko Fujisawa
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yoshimichi Okayama
- grid.260969.20000 0001 2149 8846Allergy and Immunology Research Project Team, Research Institute of Medical Science, Center for Allergy, Center for Medical Education, Nihon University School of Medicine, Tokyo, 173-8610 Japan
| | - Jun Kunisawa
- grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan ,grid.482562.fLaboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085 Japan
| | - Masato Kubo
- grid.509459.40000 0004 0472 0267Laboratory for Cytokine Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045 Japan ,grid.143643.70000 0001 0660 6861Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Chiba, 278-0022 Japan
| | - Naoki Takemura
- grid.136304.30000 0004 0370 1101Department of Innovative Medicine and Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871 Japan
| | - Satoshi Uematsu
- grid.136304.30000 0004 0370 1101Department of Innovative Medicine and Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108–8639 Japan ,grid.261445.00000 0001 1009 6411Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585 Japan
| | - Shizuo Akira
- grid.136593.b0000 0004 0373 3971Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| | - Jiro Kitaura
- grid.258269.20000 0004 1762 2738Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421 Japan
| | - Takao Takahashi
- grid.26091.3c0000 0004 1936 9959Department of Pediatrics, Keio University School of Medicine, Tokyo, 160-8582 Japan
| | - Toshinori Nakayama
- grid.136304.30000 0004 0370 1101Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan
| | - Hiroshi Kiyono
- grid.26999.3d0000 0001 2151 536XDepartment of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.26091.3c0000 0004 1936 9959Department of Pediatrics, Keio University School of Medicine, Tokyo, 160-8582 Japan ,grid.266100.30000 0001 2107 4242Division of Gastroenterology, Department of Medicine, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA 92093-0956 USA ,grid.136304.30000 0004 0370 1101Institute for Global Prominent Research, Chiba University, Chiba, 260-8670 Japan ,grid.136304.30000 0004 0370 1101Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, 260-8670 Japan
| |
Collapse
|
35
|
Radmanesh F, Mahmoudi M, Yazdanpanah E, Keyvani V, Kia N, Nikpoor AR, Zafari P, Esmaeili SA. The immunomodulatory effects of mesenchymal stromal cell-based therapy in human and animal models of systemic lupus erythematosus. IUBMB Life 2020; 72:2366-2381. [PMID: 33006813 DOI: 10.1002/iub.2387] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune inflammatory disease with no absolute cure. Although the exact etiopathogenesis of SLE is still enigmatic, it has been well demonstrated that a combination of genetic predisposition and environmental factors trigger a disturbance in immune responses and thereby participate in the development of this condition. Almost all available therapeutic strategies in SLE are primarily based on the administration of immunosuppressive drugs and are not curative. Mesenchymal stromal cells (MSCs) are a subset of non-hematopoietic adult stem cells that can be isolated from many adult tissues and are increasingly recognized as immune response modulating agents. MSC-mediated inhibition of immune responses is a complex mechanism that involves almost every aspect of the immune response. MSCs suppress the maturation of antigen-presenting cells (DC and MQ), proliferation of T cells (Th1, T17, and Th2), proliferation and immunoglobulin production of B cells, the cytotoxic activity of CTL and NK cells in addition to increasing regulatory cytokines (TGF-β and IL10), and decreasing inflammatory cytokines (IL17, INF-ϒ, TNF-α, and IL12) levels. MSCs have shown encouraging results in the treatment of several autoimmune diseases, in particular SLE. This report aims to review the beneficial and therapeutic properties of MSCs; it also focuses on the results of animal model studies, preclinical studies, and clinical trials of MSC therapy in SLE from the immunoregulatory aspect.
Collapse
Affiliation(s)
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahideh Keyvani
- Molecular Genetics, Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadia Kia
- Skin Cancer Prevention Research Center, Torvergata University of Medical Sciences, Rome, Italy
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parisa Zafari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Science, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Science, Sari, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
Noguchi M, Hirata N, Tanaka T, Suizu F, Nakajima H, Chiorini JA. Autophagy as a modulator of cell death machinery. Cell Death Dis 2020; 11:517. [PMID: 32641772 PMCID: PMC7343815 DOI: 10.1038/s41419-020-2724-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 01/07/2023]
Abstract
The balance between cell death and survival is a critical parameter in the regulation of cells and the maintenance of homeostasis in vivo. Three major mechanisms for cell death have been identified in mammalian cells: apoptosis (type I), autophagic cell death (type II), and necrosis (type III). These three mechanisms have been suggested to engage in cross talk with each other. Among them, autophagy was originally characterized as a cell survival mechanism for amino acid recycling during starvation. Whether autophagy functions primarily in cell survival or cell death is a critical question yet to be answered. Here, we present a comprehensive review of the cell death-related events that take place during autophagy and their underlying mechanisms in cancer and autoimmune disease development.
Collapse
Affiliation(s)
- Masayuki Noguchi
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Noriyuki Hirata
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tsutomu Tanaka
- grid.94365.3d0000 0001 2297 5165National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD USA
| | - Futoshi Suizu
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Nakajima
- grid.136304.30000 0004 0370 1101Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - John A. Chiorini
- grid.94365.3d0000 0001 2297 5165National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
37
|
Garelli CJ, Refat MA, Nanaware PP, Ramirez-Ortiz ZG, Rashighi M, Richmond JM. Current Insights in Cutaneous Lupus Erythematosus Immunopathogenesis. Front Immunol 2020; 11:1353. [PMID: 32714331 PMCID: PMC7343764 DOI: 10.3389/fimmu.2020.01353] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/27/2020] [Indexed: 12/25/2022] Open
Abstract
Cutaneous Lupus Erythematosus (CLE) is a clinically diverse group of autoimmune skin diseases with shared histological features of interface dermatitis and autoantibodies deposited at the dermal-epidermal junction. Various genetic and environmental triggers of CLE promote infiltration of T cells, B cells, neutrophils, antigen presenting cells, and NK cells into lesional skin. In this mini-review, we will discuss the clinical features of CLE, insights into CLE immunopathogenesis, and novel treatment approaches.
Collapse
Affiliation(s)
- Colton J. Garelli
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Maggi Ahmed Refat
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Padma P. Nanaware
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Zaida G. Ramirez-Ortiz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Mehdi Rashighi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jillian M. Richmond
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
38
|
Brune Z, Rice MR, Barnes BJ. Potential T Cell-Intrinsic Regulatory Roles for IRF5 via Cytokine Modulation in T Helper Subset Differentiation and Function. Front Immunol 2020; 11:1143. [PMID: 32582209 PMCID: PMC7283537 DOI: 10.3389/fimmu.2020.01143] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022] Open
Abstract
Interferon Regulatory Factor 5 (IRF5) is one of nine members of the IRF family of transcription factors. Although initially discovered as a key regulator of the type I interferon and pro-inflammatory cytokine arm of the innate immune response, IRF5 has now been found to also mediate pathways involved in cell growth and differentiation, apoptosis, metabolic homeostasis and tumor suppression. Hyperactivation of IRF5 has been implicated in numerous autoimmune diseases, chief among them systemic lupus erythematosus (SLE). SLE is a heterogeneous autoimmune disease in which patients often share similar characteristics in terms of autoantibody production and strong genetic risk factors, yet also possess unique disease signatures. IRF5 pathogenic alleles contribute one of the strongest risk factors for SLE disease development. Multiple models of murine lupus have shown that loss of Irf5 is protective against disease development. In an attempt to elucidate the regulatory role(s) of IRF5 in driving SLE pathogenesis, labs have begun to examine the function of IRF5 in several immune cell types, including B cells, macrophages, and dendritic cells. A somewhat untouched area of research on IRF5 is in T cells, even though Irf5 knockout mice were reported to have skewing of T cell subsets from T helper 1 (Th1) and T helper 17 (Th17) toward T helper 2 (Th2), indicating a potential role for IRF5 in T cell regulation. However, most studies attributed this T cell phenotype in Irf5 knockout mice to dysregulation of antigen presenting cell function rather than an intrinsic role for IRF5 in T cells. In this review, we offer a different interpretation of the literature. The role of IRF5 in T cells, specifically its control of T cell effector polarization and the resultant T cell-mediated cytokine production, has yet to be elucidated. A strong understanding of the regulatory role(s) of this key transcription factor in T cells is necessary for us to grasp the full picture of the complex pathogenesis of autoimmune diseases like SLE.
Collapse
Affiliation(s)
- Zarina Brune
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Matthew R. Rice
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Betsy J. Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
39
|
Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells. Int J Mol Sci 2020; 21:ijms21072356. [PMID: 32235291 PMCID: PMC7177531 DOI: 10.3390/ijms21072356] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/12/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Regulatory T cells (Tregs) are important mediators of immunological self-tolerance and homeostasis. Being cluster of differentiation 4+Forkhead box protein3+ (CD4+FOXP3+), these cells are a subset of CD4+ T lymphocytes and can originate from the thymus (tTregs) or from the periphery (pTregs). The malfunction of CD4+ Tregs is associated with autoimmune responses such as rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), inflammatory bowel diseases (IBD), psoriasis, systemic lupus erythematosus (SLE), and transplant rejection. Recent evidence supports an opposed role in sepsis. Therefore, maintaining functional Tregs is considered as a therapy regimen to prevent autoimmunity and allograft rejection, whereas blocking Treg differentiation might be favorable in sepsis patients. It has been shown that Tregs can be generated from conventional naïve T cells, called iTregs, due to their induced differentiation. Moreover, Tregs can be effectively expanded in vitro based on blood-derived tTregs. Taking into consideration that the suppressive role of Tregs has been mainly attributed to the expression and function of the transcription factor Foxp3, modulating its expression and binding to the promoter regions of target genes by altering the chromatin histone acetylation state may turn out beneficial. Hence, we discuss the role of histone deacetylation inhibitors as epigenetic modulators of Tregs in this review in detail.
Collapse
|
40
|
Liu TT, Zeng XP, Gu ML, Deng AM. Increased CD200 levels in peripheral blood mononuclear cells of patients with primary Sjögren's syndrome. Int J Rheum Dis 2020; 23:654-660. [PMID: 32180363 DOI: 10.1111/1756-185x.13810] [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: 09/30/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease with an unknown etiology. CD200 is associated with many autoimmune diseases, but little is known about its role in pSS. This study aims to correlate the expression of CD200 with pSS and evaluate its significance. METHODS Plasma CD200, CD200R, and interleukin (IL)-17 levels were measured and analyzed by enzyme-linked immunosorbent assay. Messenger RNA levels of CD200 and CD200R in peripheral blood mononuclear cells (PBMCs) were quantified by quantitative real-time polymerase chain reaction (RT-qPCR). Following pretreatment of CD200-Fc, the protein levels of IL-17A were measured in PBMCs from patients and healthy controls. RESULTS Results showed that, compared to CD200 in healthy controls, the relative levels in PBMCs from pSS were greater than 2-fold. In addition, CD200 levels in plasma positively correlated with IL-17 levels, as well as between plasma CD200 and pSS activity indexes (including immunoglobulin G and European League Against Rheumatism SS Disease Activity Index). While CD200R levels were significantly decreased in pSS patients, no correlation could be found. Furthermore, the protein level of IL-17 decreased after pretreatment of CD200-Fc in PBMCs from pSS patients. CONCLUSION Our results suggested that the CD200/CD200R pathway is involved in pSS pathogenesis. It is hypothesized that regulation of IL-17 expression affects Th17 differentiation. This newly discovered pathway could give rise to a novel targeted therapy for pSS.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Department of Laboratory Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiang-Peng Zeng
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force (Fuzhou General Hospital of Fujian Medical University, Eastern Hospital Affiliated to Xiamen University), Fuzhou, China
| | - Ming-Li Gu
- Department of Laboratory Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - An-Mei Deng
- Department of Laboratory Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
41
|
Ferreira RC, Castro Dopico X, Oliveira JJ, Rainbow DB, Yang JH, Trzupek D, Todd SA, McNeill M, Steri M, Orrù V, Fiorillo E, Crouch DJM, Pekalski ML, Cucca F, Tree TI, Vyse TJ, Wicker LS, Todd JA. Chronic Immune Activation in Systemic Lupus Erythematosus and the Autoimmune PTPN22 Trp 620 Risk Allele Drive the Expansion of FOXP3 + Regulatory T Cells and PD-1 Expression. Front Immunol 2019; 10:2606. [PMID: 31781109 PMCID: PMC6857542 DOI: 10.3389/fimmu.2019.02606] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 02/01/2023] Open
Abstract
In systemic lupus erythematosus (SLE), perturbed immunoregulation underpins a pathogenic imbalance between regulatory and effector CD4+ T-cell activity. However, to date, the characterization of the CD4+ regulatory T cell (Treg) compartment in SLE has yielded conflicting results. Here we show that patients have an increased frequency of CD4+FOXP3+ cells in circulation owing to a specific expansion of thymically-derived FOXP3+HELIOS+ Tregs with a demethylated FOXP3 Treg-specific demethylated region. We found that the Treg expansion was strongly associated with markers of recent immune activation, including PD-1, plasma concentrations of IL-2 and the type I interferon biomarker soluble SIGLEC-1. Since the expression of the negative T-cell signaling molecule PTPN22 is increased and a marker of poor prognosis in SLE, we tested the influence of its missense risk allele Trp620 (rs2476601C>T) on Treg frequency. Trp620 was reproducibly associated with increased frequencies of thymically-derived Tregs in blood, and increased PD-1 expression on both Tregs and effector T cells (Teffs). Our results support the hypothesis that FOXP3+ Tregs are increased in SLE patients as a consequence of a compensatory mechanism in an attempt to regulate pathogenic autoreactive Teff activity. We suggest that restoration of IL-2-mediated homeostatic regulation of FOXP3+ Tregs by IL-2 administration could prevent disease flares rather than treating at the height of a disease flare. Moreover, stimulation of PD-1 with specific agonists, perhaps in combination with low-dose IL-2, could be an effective therapeutic strategy in autoimmune disease and in other immune disorders.
Collapse
Affiliation(s)
- Ricardo C. Ferreira
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Xaquin Castro Dopico
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - João J. Oliveira
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Daniel B. Rainbow
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jennie H. Yang
- Department of Immunobiology, NIHR Biomedical Research Centre, King's College London, London, United Kingdom
| | - Dominik Trzupek
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Sarah A. Todd
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Mhairi McNeill
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Daniel J. M. Crouch
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Marcin L. Pekalski
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Tim I. Tree
- Department of Immunobiology, NIHR Biomedical Research Centre, King's College London, London, United Kingdom
| | - Tim J. Vyse
- Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - Linda S. Wicker
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - John A. Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Wellcome Centre for Human Genetics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The management of lupus nephritis remains unsatisfactory due to insufficiently effective treatment regimens and the dearth of reliable predictors of disease onset or progression to guide individualized therapeutic decisions. This review summarizes new findings related to lupus nephritis over the last 18 months and discusses clinical needs that should be considered to advance trials of mechanism-based therapeutic strategies. RECENT FINDINGS Collaborative teams are addressing how to improve disease definitions and are developing predictive models for disease onset, disease response and risk of flare in individual patients. More attention is being paid to clinical trial design. Advanced technologic approaches are allowing the analysis of small amounts of human tissue and urine in unprecedented detail so as to discover new pathogenic mechanisms and identify disease biomarkers. Novel therapies continue to be tested in disease models and include new strategies to protect renal tissue from cell damage and fibrosis. SUMMARY The collaborative efforts of patients, clinical and translational researchers, the pharmaceutical industry and funding sources are needed to advance therapies for lupus nephritis. Specialized clinical centers can then deliver optimal and more personalized patient care that will improve patient outcomes.
Collapse
Affiliation(s)
- Anne Davidson
- Center for Autoimmunity, Musculoskeletal and Hematologic Diseases, Feinstein Institute for Medical Research, New York, New York, USA
| | | | | |
Collapse
|
43
|
Chu Y, Zhao C, Zhang B, Wang X, Wang Y, An J, Chen J. Restoring T-helper 17 cell/regulatory T-cell balance and decreasing disease activity by rapamycin and all-trans retinoic acid in patients with systemic lupus erythematosus. Lupus 2019; 28:1397-1406. [PMID: 31551029 DOI: 10.1177/0961203319877239] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE This study aimed to investigate the effect of rapamycin (RAPA) alone or in combination with all-trans retinoic acid (ATRA) on the T-helper 17 (Th17) cell/regulatory T-cell (Treg) balance in patients with systemic lupus erythematosus (SLE) and to evaluate the clinical efficacy. METHODS Seventy patients with SLE were enrolled. They were randomly and equally divided into RAPA and RAPA + ATRA groups. The number of Th17 and Treg cells was measured by flow cytometry before and after treatment for 6, 12 and 24 weeks. The SLE Disease Activity Index (SLEDAI) score and the prednisone dose before and after treatment were used to evaluate the efficacy between the two groups. RESULTS In both groups, at different time points after treatment, the number of Th17 cells (p = 0.003) and Th17/Treg ratio (p = 0.044) reduced, while the number of Treg cells (p = 0.574) tended to increase. The SLEDAI score and the dose of prednisone decreased significantly (p < 0.001). There was no significant difference in the number of Th17 cells (p = 0.089), Treg cells (p = 0.059), Th17/Treg ratio (p = 0.580), SLEDAI score (p = 0.127) and the dose of prednisone (p = 0.329) between the two groups. CONCLUSION Disease activity in SLE patients reduced with RAPA alone or in conjunction with ATRA, reducing glucocorticoid requirement. One of its mechanisms of action may be regulating the Th17/Treg cell balance, which provides a new model for the pathogenesis and potential treatment of SLE.
Collapse
Affiliation(s)
- Y Chu
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - C Zhao
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - B Zhang
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - X Wang
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Y Wang
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - J An
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - J Chen
- Department of Rheumatology and Immunology, the Second Hospital of Shanxi Medical University, Taiyuan, PR China
| |
Collapse
|
44
|
Li X, Luo F, Li J, Luo C. MiR-183 delivery attenuates murine lupus nephritis-related injuries via targeting mTOR. Scand J Immunol 2019; 90:e12810. [PMID: 31325389 DOI: 10.1111/sji.12810] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/11/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) play a vital role in the occurrence and development of many human diseases, including systemic lupus erythematosus (SLE). SLE is an autoimmune disease characterized by the production of autoantibodies against nuclear antigens and multiorgan involvement. Study of miRNAs involved in SLE provides new insights into the pathogenesis of SLE and might lead to the identification of new therapeutic interventions. The aim of this study was to investigate the effect of miR-183 injection on the progression of SLE by using MRL/lpr mouse model. The expression levels of miR-183 and mTOR mRNA were detected by quantitative real-time PCR assay. The effect of miR-183 on the course of spontaneous disease progression in the MRL/lpr mice was examined by intraperitoneal injection of miR-183 into mice and followed by monitoring lifespan, anti-dsDNA antibody levels, urinary albumin levels, blood urea nitrogen (BUN) levels, and Tregs and Th17 cell population. We found that miR-183 injection resulted in reduction of anti-DNA antibody and immune complex component levels, restoration of Tregs and Th17 cell population and prolongation of survival. Our findings suggest that miR-183 injection may serve as an effective therapeutic treatment for delaying or easing pathologic features of SLE.
Collapse
Affiliation(s)
- Xiuzhen Li
- Department of Nephrology, Liaocheng People's Hospital, Liaocheng, China
| | - Feng Luo
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Li
- Department of Nephrology, Liaocheng People's Hospital, Liaocheng, China
| | - Congjuan Luo
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
45
|
Mijnheer G, van Wijk F. T-Cell Compartmentalization and Functional Adaptation in Autoimmune Inflammation: Lessons From Pediatric Rheumatic Diseases. Front Immunol 2019; 10:940. [PMID: 31143175 PMCID: PMC6520654 DOI: 10.3389/fimmu.2019.00940] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/11/2019] [Indexed: 12/30/2022] Open
Abstract
Chronic inflammatory diseases are characterized by a disturbed immune balance leading to recurring episodes of inflammation in specific target tissues, such as the joints in juvenile idiopathic arthritis. The tissue becomes infiltrated by multiple types of immune cell, including high numbers of CD4 and CD8 T-cells, which are mostly effector memory cells. Locally, these T-cells display an environment-adapted phenotype, induced by inflammation- and tissue-specific instructions. Some of the infiltrated T-cells may become tissue resident and play a role in relapses of inflammation. Adaptation to the environment may lead to functional (re)programming of cells and altered cellular interactions and responses. For example, specifically at the site of inflammation both CD4 and CD8 T-cells can become resistant to regulatory T-cell-mediated regulation. In addition, CD8 and CD4 T-cells show a unique profile with pro- and anti-inflammatory features coexisting in the same compartment. Also regulatory T-cells are neither homogeneous nor static in nature and show features of functional differentiation, and plasticity in inflammatory environments. Here we will discuss the recent insights in T-cell functional specialization, regulation, and clonal expansion in local (tissue) inflammation.
Collapse
Affiliation(s)
- Gerdien Mijnheer
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Femke van Wijk
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
46
|
Autophagy in regulatory T cells: A double-edged sword in disease settings. Mol Immunol 2019; 109:43-50. [PMID: 30852245 DOI: 10.1016/j.molimm.2019.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/03/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is an evolutionarily conserved catabolic process that directs cytoplasmic proteins, organelles and microbes to lysosomes for degradation. It not only represents an essential cell-intrinsic mechanism to protect against internal and external stresses but also shapes both innate and adaptive immunity. Regulatory T cells (Tregs) are a developmentally and functionally distinct T cell subpopulation engaged in sustaining immunological self-tolerance and homeostasis. There is compelling evidence that autophagy is actively regulated in Tregs and serves as a central signal-dependent controller for Tregs by restraining excessive apoptotic and metabolic activities. In this review, we discuss how autophagy modulates the stability and functionality of Tregs in different disease settings, and provide a perspective on how manipulation of autophagy enables better control of immune response by targeting the generation of Tregs and the maintenance of their stability.
Collapse
|
47
|
Toomer KH, Lui JB, Altman NH, Ban Y, Chen X, Malek TR. Essential and non-overlapping IL-2Rα-dependent processes for thymic development and peripheral homeostasis of regulatory T cells. Nat Commun 2019; 10:1037. [PMID: 30833563 PMCID: PMC6399264 DOI: 10.1038/s41467-019-08960-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/11/2019] [Indexed: 01/06/2023] Open
Abstract
IL-2R signaling is essential for regulatory T cell (Treg) function. However, the precise contribution of IL-2 during Treg thymic development, peripheral homeostasis and lineage stability remains unclear. Here we show that IL-2R signaling is required by thymic Tregs at an early step for expansion and survival, and a later step for functional maturation. Using inducible, conditional deletion of CD25 in peripheral Tregs, we also find that IL-2R signaling is indispensable for Treg homeostasis, whereas Treg lineage stability is largely IL-2-independent. CD25 knockout peripheral Tregs have increased apoptosis, oxidative stress, signs of mitochondrial dysfunction, and reduced transcription of key enzymes of lipid and cholesterol biosynthetic pathways. A divergent IL-2R transcriptional signature is noted for thymic Tregs versus peripheral Tregs. These data indicate that IL-2R signaling in the thymus and the periphery leads to distinctive effects on Treg function, while peripheral Treg survival depends on a non-conventional mechanism of metabolic regulation. Interleukin-2 (IL-2) signaling is required for regulatory T (Treg) cell differentiation in the thymus, but its function in peripheral Tregs is still unclear. Here the authors show, using inducible deletion of IL-2 receptor subunit CD25, that IL-2 signaling is essential for maintaining peripheral Treg homeostasis, but dispensable for lineage stability.
Collapse
Affiliation(s)
- Kevin H Toomer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Jen Bon Lui
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Norman H Altman
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Yuguang Ban
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Xi Chen
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA. .,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
48
|
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
|
49
|
Treg/Th17 imbalance is associated with poor autoimmune hepatitis prognosis. Clin Immunol 2019; 198:79-88. [DOI: 10.1016/j.clim.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/04/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
|
50
|
Moulton VR. Sex Hormones in Acquired Immunity and Autoimmune Disease. Front Immunol 2018; 9:2279. [PMID: 30337927 PMCID: PMC6180207 DOI: 10.3389/fimmu.2018.02279] [Citation(s) in RCA: 375] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022] Open
Abstract
Women have stronger immune responses to infections and vaccination than men. Paradoxically, the stronger immune response comes at a steep price, which is the high incidence of autoimmune diseases in women. The reasons why women have stronger immunity and higher incidence of autoimmunity are not clear. Besides gender, sex hormones contribute to the development and activity of the immune system, accounting for differences in gender-related immune responses. Both innate and adaptive immune systems bear receptors for sex hormones and respond to hormonal cues. This review focuses on the role of sex hormones particularly estrogen, in the adaptive immune response, in health, and autoimmune disease with an emphasis on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|