1
|
Zhao J, Peng W, Wu S, Wang W. Evaluation of disease activity in systemic lupus erythematosus using standard deviation of lymphocyte volume combined with red blood cell count and lymphocyte percentage. Sci Rep 2024; 14:22470. [PMID: 39341869 PMCID: PMC11439007 DOI: 10.1038/s41598-024-72977-w] [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: 06/21/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Systemic lupus erythematosus (SLE) commonly damages the blood system and often manifests as blood cell abnormalities. The performance of biomarkers for predicting SLE activity still requires further improvement. This study aimed to analyze blood cell parameters to identify key indicators for a SLE activity prediction model. Clinical data of 138 patients with SLE (high activity, n = 40; moderate activity, n = 44; mild activity, n = 37; low activity, n = 17) and 100 healthy controls (HCs) were retrospectively analyzed. Data from 89 paired admission-discharge patients with SLE were collected. Differences and associations between blood cell parameters and disease indicators, as well as the relationship between the these parameters and organ damage, were examined. Machine-learning methods were employed to develop a prediction model for disease activity evaluation. Most blood cell parameters (22/26, 84.62%) differed significantly between patients with SLE and HCs. Analysis of 89 paired patients with SLE revealed significant changes in most blood cell parameters at discharge. The standard deviation of lymphocyte volume (SD-V-LY), red blood cell (RBC) count, lymphocyte percentage (LY%), hemoglobin(HGB), hematocrit(HCT), and neutrophil percentage(NE%) correlated with disease activity. By employing machine learning, an optimal model was established to predict active SLE using SD-V-LY, RBC count, and LY% (area under the curve [AUC] = 0.908, sensitivity = 0.811). External validation indicated impressive performance (AUC = 0.940, sensitivity = 0.833). Correlation analysis revealed that SD-V-LY was positively correlated with ESR, IgG, IgA, and IgM but was negatively correlated with C3 and C4. The RBC count was linked to renal and hematopoietic system impairments, whereas LY% was associated with joint/muscle involvement. In conclusion, SD-V-LY is associated with SLE disease activity. SD-V-LY combined with RBC count and LY% contributes to a prediction model, which can be utilized as an effective tool for assessing SLE activity.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Wanchan Peng
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Siyu Wu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Wei Wang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Peng L, Wang P, Xu X, Chen D, Xu F, Yang F, Yang S, Xia H, Liu ZH, Qin W. Inhibition of receptor interacting protein kinase-1 (RIPK1) in the treatment of murine lupus. Lupus Sci Med 2024; 11:e001146. [PMID: 38906550 PMCID: PMC11191810 DOI: 10.1136/lupus-2024-001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a type of autoimmune disease that involves multiple organs involved as well as cytokine dysregulation. The treatment of SLE is still challenging due to the side effects of the different drugs used. Receptor-interacting protein kinase 1 (RIPK1) is a kinase involved in T cell homeostasis and autoinflammation. Although clinical trials have shown that RIPK1 inhibition exhibits significant efficacy in different autoimmune diseases, its role in SLE remains unclear. METHODS MRL/lpr lupus-prone mice received RIPK1 inhibitor ZJU37 or vehicle intraperitoneally for 10 weeks. A BM12-induced chronic graft-versus-host-disease (cGVHD) lupus-like model was introduced in RIPK1 D138N mice or C57BL/6 mice. Nephritis, serum autoantibody levels, dysregulation of adaptive immune response and cytokines were compared in treated and untreated mice. RESULTS ZJU37 alleviated the clinical features of the MRL/lpr mice including nephritis and anti-dsDNA antibody production. In addition, ZJU37 treatment reduced the proportion of double-negative T cells in the spleen and the cytokines of TNFα, IFN-γ, IL-6, IL-17 and IL-1β in the serum. Moreover, RIPK1 D138N mice were able to prevent the cGVHD lupus-like model from SLE attack, manifesting as anti-dsDNA antibody production, the proliferation of germinal centre B cells, plasma cells, and T follicular helper cells as well as IgG and C3 deposits in kidneys. CONCLUSION RIPK1 inhibition has a protective effect in the mouse model of SLE and can potentially become a new therapeutic target for SLE in humans.
Collapse
Affiliation(s)
- Lin Peng
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Pengcheng Wang
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xiaodong Xu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Dacheng Chen
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Feng Xu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Fan Yang
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Shuying Yang
- Department of Biochemistry and Molecular Medical Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hongguang Xia
- Department of Biochemistry and Molecular Medical Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhi-Hong Liu
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Weisong Qin
- National Clinical Research Center for Kidney Disease, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Zhao X, Ji W, Lu Y, Liu W, Guo F. Triptolide regulates the balance of Tfr/Tfh in lupus mice. Adv Rheumatol 2023; 63:29. [PMID: 37408016 DOI: 10.1186/s42358-023-00311-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/15/2023] [Indexed: 07/07/2023] Open
Abstract
INTRODUCTION/OBJECTIVES Systemic lupus erythematosus (SLE) is a classic prototype of the multisystem autoimmune disease and follows a relapsing and remitting course. Triptolide is a diterpene triepoxide extracted from Chinese medicine Tripterygium wilfordii Hook F, with potent immunosuppressive and anti-inflammatory properties. Our previous work observed that triptolide alleviated lupus in MRL/lpr lupus mice with the upregulation of regulatory T cells (Treg) proportion in previous study. In this study, we explored the proportion of follicular T regulatory (Tfr), follicular T helper (Tfh) and germinal center (GC) B cells in lupus mice and evaluated the efficacy of triptolide for lupus treatment in vivo. METHODS 20 female MRL/lpr mice were randomly divided into 2 treatment groups and treated orally with vehicle or triptolide. C3H mice were all housed as controlled group and treated orally with vehicle. The percentage of Tfr cells, Tfh cells and GC B cells in spleen of mice were detected by Flow cytometric analysis and immunohistochemistry after 13 weeks of treatment. RESULTS We found that the percentage of Tfr cells decreased in MRL/lpr mice compared with controlled mice. The percentage of Tfh cells in MRL/lpr mice was significantly higher compared with that in controlled mice. The ratio of Tfr/Tfh is also decreased in lupus mice. After treated with triptolide in MRL/Lpr mice in vivo, the percentage of Tfr cells and ratio of Tfr/Tfh increased. The proportion of GC B cells also decreased in mice treated with triptolide by FACS and immunohistochemistry. CONCLUSIONS Our results demonstrate that the effect of triptolide in alleviating lupus is partly by reversing immune imbalance with increased percentage of Tfr cells and ratio of Tfr/Tfh. Triptolide might also has effect on immune response through inhibiting proliferating GC B cells.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210027, China
| | - Wei Ji
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210027, China
| | - Yan Lu
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210027, China
| | - Weiwei Liu
- Department of Medical Examination Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210027, China
| | - Feng Guo
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210027, China.
| |
Collapse
|
4
|
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: 0.5] [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
|
5
|
Akama-Garren EH, Carroll MC. Lupus Susceptibility Loci Predispose Mice to Clonal Lymphocytic Responses and Myeloid Expansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2403-2424. [PMID: 35477687 PMCID: PMC9254690 DOI: 10.4049/jimmunol.2200098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 05/17/2023]
Abstract
Lupus susceptibility results from the combined effects of numerous genetic loci, but the contribution of these loci to disease pathogenesis has been difficult to study due to the large cellular heterogeneity of the autoimmune immune response. We performed single-cell RNA, BCR, and TCR sequencing of splenocytes from mice with multiple polymorphic lupus susceptibility loci. We not only observed lymphocyte and myeloid expansion, but we also characterized changes in subset frequencies and gene expression, such as decreased CD8 and marginal zone B cells and increased Fcrl5- and Cd5l-expressing macrophages. Clonotypic analyses revealed expansion of B and CD4 clones, and TCR repertoires from lupus-prone mice were distinguishable by algorithmic specificity prediction and unsupervised machine learning classification. Myeloid differential gene expression, metabolism, and altered ligand-receptor interaction were associated with decreased Ag presentation. This dataset provides novel mechanistic insight into the pathophysiology of a spontaneous model of lupus, highlighting potential therapeutic targets for autoantibody-mediated disease.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA; and
- Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA; and
| |
Collapse
|
6
|
Sonoda S, Yamaza T. A New Target of Dental Pulp-Derived Stem Cell-Based Therapy on Recipient Bone Marrow Niche in Systemic Lupus Erythematosus. Int J Mol Sci 2022; 23:ijms23073479. [PMID: 35408840 PMCID: PMC8998830 DOI: 10.3390/ijms23073479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/04/2023] Open
Abstract
Recent advances in mesenchymal stem/stromal cell (MSC) research have led us to consider the feasibility of MSC-based therapy for various diseases. Human dental pulp-derived MSCs (hDPSCs) have been identified in the dental pulp tissue of deciduous and permanent teeth, and they exhibit properties with self-renewal and in vitro multipotency. Interestingly, hDPSCs exhibit superior immunosuppressive functions toward immune cells, especially T lymphocytes, both in vitro and in vivo. Recently, hDPSCs have been shown to have potent immunomodulatory functions in treating systemic lupus erythematosus (SLE) in the SLE MRL/lpr mouse model. However, the mechanisms underlying the immunosuppressive efficacy of hDPSCs remain unknown. This review aims to introduce a new target of hDPSC-based therapy on the recipient niche function in SLE.
Collapse
|
7
|
Zhang J, Chang L, Sun Y, Qin M, Wang X, Guo Y. Disabled-2 (DAB2) overexpression mediates immune suppression in systemic lupus erythematosus by modulating Treg/Th17 cell differentiation. Clin Exp Pharmacol Physiol 2022; 49:596-607. [PMID: 35108421 DOI: 10.1111/1440-1681.13630] [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: 07/28/2021] [Revised: 12/27/2021] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder. T helper 17 (Th17) and regulatory T (Treg) cells play key roles in SLE progression. Disabled-2 (DAB2) exhibits immunomodulatory effects in inflammatory diseases. However, the role of DAB2 in SLE and the precise mechanisms remain unknown. Herein, a decreased DAB2 expression and an increased miR-448-3p level were observed in PBMCs from SLE patients. DAB2 level was negatively correlated with SLE Disease Activity Index (SLEDAI), suggesting a functional correlation between DAB2 and SLE. To test this, we employed 8-week-old MRL/lpr mice and treated them with lentivirus-mediated DAB2 or its negative control (LV-NC). LV-DAB2 treatment increased DAB2 expression and reduced serum IgG and anti-dsDNA IgG levels. DAB2 up-regulation alleviated splenomegaly and lymphadenopathy and SLE-related organ damage. Moreover, DAB2 enhanced the percentage of CD25+ Foxp3+ Treg cells but reduced Th17 cell frequency in lupus, along with the reduction in TNF-α, IL-6 and IL-17A levels, and the elevation in IL-10. In vitro, naive CD4+ T cells isolated from MRL/lpr mice were polarized into Th17 or Treg phenotypes and treated with lentivirus. LV-DAB2 treatment down-regulated IL-17A expression and inhibited the generation of CD4+ IL-17A+ Th17 cells. Also, DAB2 triggered the production of IL-10 and the activation of Treg cells. Furthermore, DAB2 was verified as a direct target for miR-448-3p. MiR-448-3p overexpression canceled the promoting effect of DAB2 on Treg cell differentiation. Taken together, DAB2 exerts an immunosuppressive effect on SLE through promoting Treg cell activation and inhibiting Th17 cell differentiation, which may be modulated by miR-448-3p.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Lihua Chang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Yue Sun
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Muting Qin
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Xiaofei Wang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Yun Guo
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| |
Collapse
|
8
|
Long Y, Li W, Feng J, Ma Y, Sun Y, Xu L, Song Y, Liu C. Follicular helper and follicular regulatory T cell subset imbalance is associated with higher activated B cells and abnormal autoantibody production in primary anti-phospholipid syndrome patients. Clin Exp Immunol 2021; 206:141-152. [PMID: 34309827 PMCID: PMC8506124 DOI: 10.1111/cei.13647] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/07/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Primary anti-phospholipid antibody syndrome (pAPS) is a multi-organ autoimmune disease, and autoantibodies are involved in its pathogenesis. Follicular helper T cells (Tfh) and follicular regulatory T cells (Tfr) are critical for B cell maturation and antibody production, but their roles in pAPS remain unknown. We enrolled 32 pAPS patients and 23 healthy controls (HCs) and comprehensively analyzed circulating Tfh and Tfr, as well as their subsets, using flow cytometry. Clinical data including autoantibody levels were collected and their correlations with Tfh and Tfr subsets were analyzed. In addition, correlation analyses between B cell functional subsets and Tfh and Tfr were performed. Changes and potential effects of serum cytokines on Tfr and Tfh were further explored. We found the circulating Tfr was significantly decreased while Tfh and Tfh/Tfr ratios were increased in pAPS patients. Tfh2, inducible T cell co-stimulator (ICOS)+ programmed cell death 1 (PD-1)+ Tfh and Ki-67+ Tfh percentages were elevated, while CD45RA- forkhead box protein 3 (FoxP3)hi , Helios+ , T cell immunoglobulin and ITIM (TIGIT)+ and Ki-67+ Tfr percentages were decreased in pAPS patients. New memory B cells and plasmablasts were increased and altered B cell subsets and serum autoantibodies were positively correlated with Tfh, Tfh2, ICOS+ PD-1+ Tfh cells and negatively associated with Tfr, CD45RA- FoxP3hi Tfr and Helios+ Tfr cells. In addition, pAPS with LA/aCL/β2GPI autoantibodies showed lower functional Tfr subsets and higher activated Tfh subsets. Serum interleukin (IL)-4, IL-21, IL-12 and transforming growth factor (TGF)-β1 were up-regulated and associated with Tfh and Tfr subset changes. Our study demonstrates that imbalance of circulating Tfr and Tfh, as well as their functional subsets, is associated with abnormal autoantibody levels in pAPS, which may contribute to the pathogenesis of pAPS.
Collapse
Affiliation(s)
- Yan Long
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Wenyi Li
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Jinghong Feng
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Yinting Ma
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Yuanyuan Sun
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Lijuan Xu
- Department of ImmunologySchool of Basic Medical SciencesPeking University Health Science CentreBeijingChina
| | - Ying Song
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| | - Chen Liu
- Department of Clinical LaboratoryPeking University People’s HospitalBeijingChina
| |
Collapse
|
9
|
Hiramatsu-Asano S, Sunahori-Watanabe K, Zeggar S, Katsuyama E, Mukai T, Morita Y, Wada J. Deletion of Mir223 Exacerbates Lupus Nephritis by Targeting S1pr1 in Faslpr/lpr Mice. Front Immunol 2021; 11:616141. [PMID: 33574820 PMCID: PMC7871001 DOI: 10.3389/fimmu.2020.616141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/09/2020] [Indexed: 12/29/2022] Open
Abstract
Objective The micro RNAs (miRNAs) and their target mRNAs are differentially expressed in various immune-mediated cells. Here, we investigated the role of Mir223 and sphingosine-1-phosphate receptor 1 (S1pr1) in the pathogenesis of systemic lupus erythematosus. Methods We analyzed miRNA and mRNA profiling data of CD4+ splenic T cells derived from MRL/MpJ-Faslpr /J mice. We performed 3' untranslated region (UTR) luciferase reporter gene assay using human umbilical vein endothelial cells (HUVECs). We generated the B6-Mir223-/-Faslpr/lpr mice and the lupus phenotypes were analyzed. Results In CD4+ splenic T cells, we identified upregulation of miR-223-3p and downregulation of the possible target, S1pr1 by RNA sequencing of MRL/MpJ-Faslpr /J mice. The transfection with miR-223-3p mimic significantly suppressed a luciferase activity in HUVEC treated with a Lentivirus vector containing 3' UTR of S1pr1. The mRNA levels of S1pr1 were significantly decreased after miR-223-3p overexpression. In B6-Mir223-/-Faslpr/lpr mice, the proportion of CD3+ T cells, CD3+CD4-CD8- cells, B cells, plasma cells, and S1PR1+CD4+ T cells in the spleen was significantly increased compared with that in B6-Mir223+/+Faslpr/lpr mice by flow cytometry. B6-Mir223-/-Faslpr/lpr mice demonstrated the elevation of glomerular and renal vascular scores associated with enhanced intraglomerular infiltration of S1PR1+CD4+ T cells. Conclusion Unexpectedly, the deletion of Mir223 exacerbated the lupus phenotypes associated with increased population of S1PR1+CD4+ T in spleen and the enhanced infiltration of S1PR1+CD4+ T cells in inflamed kidney tissues, suggesting compensatory role of Mir223 in the pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Sumie Hiramatsu-Asano
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Katsue Sunahori-Watanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Sonia Zeggar
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Eri Katsuyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoyuki Mukai
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Yoshitaka Morita
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
10
|
De Groof A, Ducreux J, Aleva F, Long AJ, Ferster A, van der Ven A, van de Veerdonk F, Houssiau FA, Lauwerys BR. STAT3 phosphorylation mediates the stimulatory effects of interferon alpha on B cell differentiation and activation in SLE. Rheumatology (Oxford) 2020; 59:668-677. [PMID: 31504941 DOI: 10.1093/rheumatology/kez354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/05/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Type I IFNs play a well-known role in the pathogenesis of SLE, through activation of CD4 T and antigen-presenting cells. Here, we investigated the effects of IFN alpha (IFNα) on SLE B cell activation and differentiation. METHODS Peripheral blood mononuclear cells (PBMCs) and purified total or naïve B cells were obtained from healthy controls and SLE patients. The effects of IFNα on B cell differentiation were studied by flow cytometry. The role of STAT3 in B cell responses to IFNα was studied using pharmacological inhibitors and PBMCs from STAT3-deficient individuals. RESULTS Incubation of normal PBMCs with IFNα induces a B cell differentiation pattern as observed spontaneously in SLE PBMCs. IFNα displays direct stimulatory effects on purified naïve B cells from healthy individuals, as evidenced by a significant induction of cell surface CD38 and CD95 in the presence of the cytokine. In purified naïve B cells, IFNα also induces STAT3 phosphorylation. IFNα-induced naïve B cell differentiation in total PBMCs is significantly inhibited in the presence of STAT3 inhibitors, or in PBMCs from individuals with STAT3 loss of function mutations. Spontaneous levels of STAT3, but not STAT1, phosphorylation are significantly higher in total B cells from SLE patients compared with controls. Pharmacological STAT3 inhibition in SLE PBMCs inhibits naïve B cell activation and differentiation. CONCLUSION IFNα displays direct stimulatory effects on B cell differentiation and activation in SLE. STAT3 phosphorylation mediates the effects of IFNα stimulation in naïve B cells, an observation that opens new therapeutic perspectives in SLE.
Collapse
Affiliation(s)
- Aurélie De Groof
- Pôle de pathologies rhumatismales inflammatoires et systémiques, Institut de Recherche Expérimentale et Cliniques, Université catholique de Louvain, Brussels, Belgium
| | - Julie Ducreux
- Pôle de pathologies rhumatismales inflammatoires et systémiques, Institut de Recherche Expérimentale et Cliniques, Université catholique de Louvain, Brussels, Belgium
| | - Floor Aleva
- Department of General Internal Medicine, Radboud University, Nijmegen, The Netherlands
| | - Andrew J Long
- Department of Pharmacology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Alina Ferster
- Service d'Onco-Hématologie, Hôpital Reine Fabiola, Brussels, Belgium
| | - Andre van der Ven
- Department of General Internal Medicine, Radboud University, Nijmegen, The Netherlands
| | - Frank van de Veerdonk
- Department of General Internal Medicine, Radboud University, Nijmegen, The Netherlands
| | - Frédéric A Houssiau
- Pôle de pathologies rhumatismales inflammatoires et systémiques, Institut de Recherche Expérimentale et Cliniques, Université catholique de Louvain, Brussels, Belgium.,Service de Rhumatologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bernard R Lauwerys
- Pôle de pathologies rhumatismales inflammatoires et systémiques, Institut de Recherche Expérimentale et Cliniques, Université catholique de Louvain, Brussels, Belgium.,Service de Rhumatologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
11
|
Zhang J, Chen C, Fu H, Yu J, Sun Y, Huang H, Tang Y, Shen N, Duan Y. MicroRNA-125a-Loaded Polymeric Nanoparticles Alleviate Systemic Lupus Erythematosus by Restoring Effector/Regulatory T Cells Balance. ACS NANO 2020; 14:4414-4429. [PMID: 32203665 DOI: 10.1021/acsnano.9b09998] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Systemic lupus erythematosus (SLE), a common lethal autoimmune disease, is characterized by effector/regulatory T cells imbalance. Current therapies are either inefficient or have severe side effects. MicroRNA-125a (miR-125a) can stabilize Treg-mediated self-tolerance by targeting effector programs, but it is significantly downregulated in peripheral T cells of patients with SLE. Therefore, overexpression of miR-125a may have therapeutic potential to treat SLE. Considering the stability and targeted delivery of miRNA remains a major challenge in vivo, we constructed a monomethoxy (polyethylene glycol)-poly(d,l-lactide-co-glycolide)-poly(l-lysine) (mPEG-PLGA-PLL) nanodelivery system to deliver miR-125a into splenic T cells. Results demonstrate that miR-125a-loaded mPEG-PLGA-PLL (PEALmiR-125a) nanoparticles (NPs) exhibit good biocompatibility and protect miR-125a from degradation, thereby prolonging the circulatory time of miRNA in vivo. In addition, PEALmiR-125a NPs are preferentially enriched in a pathological spleen and efficiently deliver miR-125a into the splenic T cells in SLE mice models. The PEALmiR-125a NPs treatment significantly alleviates SLE disease progression by reversing the imbalance of effector/regulatory T cells. Collectively, the PEALmiR-125a NPs show excellent therapeutic efficacy and safety, which may provide an effective treatment for SLE.
Collapse
Affiliation(s)
- Jiali Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Chuanrong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences (SIBS), University of Chinese Academy of Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, United States
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| |
Collapse
|
12
|
Ren J, Catalina MD, Eden K, Liao X, Read KA, Luo X, McMillan RP, Hulver MW, Jarpe M, Bachali P, Grammer AC, Lipsky PE, Reilly CM. Selective Histone Deacetylase 6 Inhibition Normalizes B Cell Activation and Germinal Center Formation in a Model of Systemic Lupus Erythematosus. Front Immunol 2019; 10:2512. [PMID: 31708928 PMCID: PMC6823248 DOI: 10.3389/fimmu.2019.02512] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/08/2019] [Indexed: 01/25/2023] Open
Abstract
Autoantibody production by plasma cells (PCs) plays a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). The molecular pathways by which B cells become pathogenic PC secreting autoantibodies in SLE are incompletely characterized. Histone deactylase 6 (HDAC6) is a unique cytoplasmic HDAC that modifies the interaction of a number of tubulin- associated proteins; inhibition of HDAC6 has been shown to be beneficial in murine models of SLE, but the downstream pathways accounting for the therapeutic benefit have not been clearly delineated. In the current study, we sought to determine whether selective HDAC6 inhibition would abrogate abnormal B cell activation in SLE. We treated NZB/W lupus mice with the selective HDAC6 inhibitor, ACY-738, for 4 weeks beginning at 20 weeks-of age. After only 4 weeks of treatment, manifestation of lupus nephritis (LN) were greatly reduced in these animals. We then used RNAseq to determine the genomic signatures of splenocytes from treated and untreated mice and applied computational cellular and pathway analysis to reveal multiple signaling events associated with B cell activation and differentiation in SLE that were modulated by HDAC6 inhibition. PC development was abrogated and germinal center (GC) formation was greatly reduced. When the HDAC6 inhibitor-treated lupus mouse gene signatures were compared to human lupus patient gene signatures, the results showed numerous immune, and inflammatory pathways increased in active human lupus were significantly decreased in the HDAC6 inhibitor treated animals. Pathway analysis suggested alterations in cellular metabolism might contribute to the normalization of lupus mouse spleen genomic signatures, and this was confirmed by direct measurement of the impact of the HDAC6 inhibitor on metabolic activities of murine spleen cells. Taken together, these studies show HDAC6 inhibition decreases B cell activation signaling pathways and reduces PC differentiation in SLE and suggest that a critical event might be modulation of cellular metabolism.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michelle D Catalina
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Kristin Eden
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Kaitlin A Read
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.,Virginia Tech Carilion Research Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xin Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Ryan P McMillan
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Matthew Jarpe
- Regenacy Pharmaceuticals, Waltham, MA, United States
| | | | - Amrie C Grammer
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Peter E Lipsky
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Christopher M Reilly
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.,Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| |
Collapse
|
13
|
Abstract
Systemic lupus erythematosus (SLE) is a devastating and heterogeneous autoimmune disease that affects multiple organs, and for which the underlying causes are unknown. The majority of SLE patients produce autoantibodies, have increased levels of type-I inflammatory cytokines, and can develop glomerulonephritis. Recent studies indicate an unexpected but strong association between increased disease activity in SLE patients and the expression of the DNA-binding protein ARID3a (A + T rich interaction domain protein 3a) in a number of peripheral blood cell types. ARID3a expression was first associated with autoantibody production in B cells; however, more recent findings also indicate associations with expression of the inflammatory cytokine interferon alpha in SLE plasmacytoid dendritic cells and low-density neutrophils. In addition, ARID3a is expressed in hematopoietic stem cells and some adult kidney progenitor cells. SLE cells expressing enhanced ARID3a levels show differential gene expression patterns compared with homologous healthy control cells, identifying new pathways potentially regulated by ARID3a. The associations of ARID3a expression with increased disease severity in SLE, suggest that it, or its downstream targets, may provide new therapeutic targets for SLE.
Collapse
|
14
|
Ma Y, Xu X, Li M, Cai J, Wei Q, Niu H. Gut microbiota promote the inflammatory response in the pathogenesis of systemic lupus erythematosus. Mol Med 2019; 25:35. [PMID: 31370803 PMCID: PMC6676588 DOI: 10.1186/s10020-019-0102-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a chronic autoimmune disease whose onset and progression are affected by genetic and environmental factors. The purpose of this study is to identify the influence of gut microbiota in the pathogenesis of SLE, and to investigate the mechanism involved. METHODS Fecal microbiota from C57/BL6 mice and SLE prone mice were examined using next-generation sequencing (NGS). Germ free mice were given fecal microbiota transplantation (FMT), and their gut microbiome and gene expression in recipients' colons were examined by NGS. The anti-double stranded DNA (anti-dsDNA) antibodies in recipients were determined using an enzyme-linked immunosorbent assay (ELISA). The immune cell profiles of mice were analyzed by flow cytometry at the 3rd week after FMT, and the expression of genes associated with SLE after FMT was determined using quantitative real-time PCR (qRT-PCR). RESULTS The fecal microbiota of SLE mice had lower community richness and diversity than healthy mice. Fecal microbiota of recipient mice were similar to their donors. Fecal microbiome from SLE mice could lead to a significant increase of anti-dsDNA antibodies and promote the immune response in recipient mice. Our results also indicated that fecal microbiome from SLE mice resulted in significant changes in the distribution of immune cells and upregulated expression of certain lupus susceptibility genes. CONCLUSIONS SLE is associated with alterations of gut microbiota. Fecal microbiome from SLE mice can induce the production of anti-dsDNA antibodies in germ free mice and stimulate the inflammatory response, and alter the expression of SLE susceptibility genes in these mice.
Collapse
Affiliation(s)
- Yiyangzi Ma
- NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaoxue Xu
- Department of Core Facility Center, Capital Medical University, Beijing, China
| | - Mengtao Li
- Division of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Cai
- Hypertension Center, Fuwai hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Disease of China, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Qiang Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Haitao Niu
- NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
15
|
Pacheco Y, Acosta-Ampudia Y, Monsalve DM, Chang C, Gershwin ME, Anaya JM. Bystander activation and autoimmunity. J Autoimmun 2019; 103:102301. [PMID: 31326230 DOI: 10.1016/j.jaut.2019.06.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 12/18/2022]
Abstract
The interaction over time of genetic, epigenetic and environmental factors (i.e., autoimmune ecology) increases or decreases the liability an individual would have to develop an autoimmune disease (AD) depending on the misbalance between risk and protective effects. Pathogens have been the most common antecedent events studied, but multiple other environmental factors including xenobiotic chemicals, drugs, vaccines, and nutritional factors have been implicated into the development of ADs. Three main mechanisms have been offered to explain the development of autoimmunity: molecular mimicry, epitope spreading, and bystander activation. The latter is characterized by auto-reactive B and T cells that undergo activation in an antigen-independent manner, influencing the development and course of autoimmunity. Activation occurs due to a combination of an inflammatory milieu, co-signaling ligands, and interactions with neighboring cells. In this review, we will discuss the studies performed seeking to define the role of bystander activation in systemic and organ-specific ADs. In all cases, we are cognizant of individual differences between hosts and the variable latency time for clinical expression of disease, all of which have made our understanding of the etiology of loss of immune tolerance difficult and enigmatic.
Collapse
Affiliation(s)
- Yovana Pacheco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA; Pediatric Immunology and Allergy, Joe DiMaggio Children's Hospital, Hollywood, FL, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA.
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Clínica del Occidente, Bogotá, Colombia.
| |
Collapse
|
16
|
Riva F, Ponzoni M, Supino D, Bertilaccio MTS, Polentarutti N, Massara M, Pasqualini F, Carriero R, Innocenzi A, Anselmo A, Veliz-Rodriguez T, Simonetti G, Anders HJ, Caligaris-Cappio F, Mantovani A, Muzio M, Garlanda C. IL1R8 Deficiency Drives Autoimmunity-Associated Lymphoma Development. Cancer Immunol Res 2019; 7:874-885. [PMID: 31018956 DOI: 10.1158/2326-6066.cir-18-0698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 01/28/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Chronic inflammation, including that driven by autoimmunity, is associated with the development of B-cell lymphomas. IL1R8 is a regulatory receptor belonging to the IL1R family, which negatively regulates NF-κB activation following stimulation of IL1R or Toll-like receptor family members. IL1R8 deficiency is associated with the development of severe autoimmune lupus-like disease in lpr mice. We herein investigated whether concomitant exacerbated inflammation and autoimmunity caused by the deficiency of IL1R8 could recapitulate autoimmunity-associated lymphomagenesis. We thus monitored B-cell lymphoma development during the aging of IL1R8-deficient lpr mice, observing an increased lymphoid cell expansion that evolved to diffuse large B-cell lymphoma (DLBCL). Molecular and gene-expression analyses showed that the NF-κB pathway was constitutively activated in Il1r8 -/-/lpr B splenocytes. In human DLBCL, IL1R8 had reduced expression compared with normal B cells, and higher IL1R8 expression was associated with a better outcome. Thus, IL1R8 silencing is associated with increased lymphoproliferation and transformation in the pathogenesis of B-cell lymphomas associated with autoimmunity.
Collapse
Affiliation(s)
- Federica Riva
- Department of Veterinary Medicine, University of Milan, Milan, Italy.,Humanitas Research Hospital, Rozzano, Italy
| | - Maurilio Ponzoni
- Ateneo Vita-Salute and Unit of Lymphoid Malignancies, IRCCS San Raffaele Scientific Institute; Pathology Unit, San Raffaele Scientific Institute, Milano, Italy
| | | | | | | | | | | | | | - Anna Innocenzi
- Ateneo Vita-Salute and Unit of Lymphoid Malignancies, IRCCS San Raffaele Scientific Institute; Pathology Unit, San Raffaele Scientific Institute, Milano, Italy
| | | | - Tania Veliz-Rodriguez
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giorgia Simonetti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Hans-Joachim Anders
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, LMU München, Germany
| | | | - Alberto Mantovani
- Humanitas Research Hospital, Rozzano, Italy.,Humanitas University, Pieve Emanuele, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Marta Muzio
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| | - Cecilia Garlanda
- Humanitas Research Hospital, Rozzano, Italy. .,Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
17
|
CD3+ B-1a Cells as a Mediator of Disease Progression in Autoimmune-Prone Mice. Mediators Inflamm 2018; 2018:9289417. [PMID: 30670930 PMCID: PMC6323491 DOI: 10.1155/2018/9289417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022] Open
Abstract
B-1a cells are distinguishable from conventional B cells, which are designated B-2 cells, on the basis of their developmental origin, surface marker expression, and functions. In addition to the unique expression of the CD5 antigen, B-1a cells are characterized by the expression level of CD23. Although B-1a cells are considered to be independent of T cells and produce natural autoantibodies that induce the clinical manifestations of autoimmune diseases, there is much debate on the role of B-1a cells in the development of autoimmune diseases. We examined the involvement of B-1a cells in autoimmune-prone mice with the lpr gene. MRL/lpr and B6/lpr mice exhibited lupus and lymphoproliferative syndromes because of the massive accumulation of CD3+CD4-CD8-B220+ T cells. Interestingly, the B220+CD23-CD5+ (B-1a) cell population in the peripheral blood and peritoneal cavity increased with age and disease progression. Ninety percent of B-1a cells were CD3 positive (CD3+ B-1a cells) and did not produce tumor necrosis factor alpha, interferon gamma, or interleukin-10. To test the possible involvement of CD3+ B-1a cells in autoimmune disease, we tried to eliminate the peripheral cells by hypotonic shock through repeated intraperitoneal injections of distilled water. The fraction of peritoneal CD3+ B-1a cells decreased, and symptoms of the autoimmune disease were much milder in the distilled water-treated MRL/lpr mice. These results suggest that CD3+ B-1a cells could be mediators of disease progression in autoimmune-prone mice.
Collapse
|
18
|
|
19
|
Liu L, Liu Y, Yuan M, Xu L, Sun H. Elevated expression of microRNA-873 facilitates Th17 differentiation by targeting forkhead box O1 (Foxo1) in the pathogenesis of systemic lupus erythematosus. Biochem Biophys Res Commun 2017; 492:453-460. [DOI: 10.1016/j.bbrc.2017.08.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/20/2017] [Indexed: 12/17/2022]
|
20
|
Immunophenotyping As a New Tool for Classification and Monitoring of Systemic Autoimmune Diseases. Clin Rev Allergy Immunol 2017; 53:177-180. [DOI: 10.1007/s12016-017-8604-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|