1
|
Yang Y, Zhang Y, Liu K, Liu M, Zhang H, Guo M. IFI27, a potential candidate molecular marker for primary Sjogren's syndrome. Clin Rheumatol 2025:10.1007/s10067-025-07409-9. [PMID: 40146445 DOI: 10.1007/s10067-025-07409-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVE The etiology of primary Sjogren's syndrome (pSS) is complex and not completely clear. This study was to identify key genes in pSS based on Gene Expression Omnibus (GEO). METHODS We downloaded the GSE40568, GSE80805, GSE127952, and GSE164885 mRNA expression profiles from GEO. Differentially expressed genes (DEGs) analyses were carried out by using the online analysis tool GEO2R and R. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to elucidate the biological processes, molecular function, cellular component, and KEGG signaling pathways for the DEGs in salivary glands (SGs) and peripheral blood mononuclear cells (PBMCs). Genes co-expressed were found in PBMCs and SGs of pSS patients. RT-qPCR was performed for validation. Finally, clinical correlation analysis and receiver operator characteristic (ROC) curve analysis were performed. RESULTS A total of thirty-nine up-regulated and one down-regulated genes were identified in pSS SGs. GO and KEGG pathway revealed that these DEGs were related to response to virus, and type I interferon signaling pathway. It was verified that fourteen genes were up-regulated in the SGs of pSS by RT-qPCR. Twenty up-regulated genes were identified in pSS patients PBMCs. Two genes were up-regulated in SGs and PBMCs of pSS patients, including IFI27 and IFI44L. The mRNA level of IFI27 was positively correlated with the disease activity of pSS patients. Furthermore, ROC analyses proved IFI27 may have diagnostic value for pSS. CONCLUSION IFI27 might serve as a potential biomarker for the early diagnosis and therapy of pSS. Key Points • IFI27 expression was significantly increased in PBMCs and SGs of pSS patients. • IFI27 was positively correlated with disease activity in pSS patients. • IFI27 might have a good diagnostic value for pSS.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
- Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ying Zhang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Ke Liu
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Meidong Liu
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Apaer A, Shi Y, Aobulitalifu A, Wen F, Muhetaer A, Ajimu N, Sulitan M, Cheng L. Identification of potential therapeutic targets for systemic lupus erythematosus based on GEO database analysis and Mendelian randomization analysis. Front Genet 2024; 15:1454486. [PMID: 39445158 PMCID: PMC11496559 DOI: 10.3389/fgene.2024.1454486] [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: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a complex autoimmune disease. Current treatments mainly rely on immunosuppressants, which lack specificity and pose challenges during treatment. This study aims to deeply explore the molecular pathogenic mechanism of SLE through gene expression databases (GEO) and bioinformatics analysis methods, combined with Mendelian randomization analysis, to provide key clues for new therapeutic targets. Methods In this study, the SLE-related gene chip dataset GSE65391 was selected from the GEO database, and the data were preprocessed and statistically analyzed using R language and bioinformatics tools. Differential expression analysis, weighted gene co-expression network analysis (WGCNA), GO, and KEGG enrichment analysis were used to screen differentially expressed genes (DEGs) for functional annotation and pathway localization. Furthermore, Mendelian randomization analysis was conducted to identify core genes closely related to SLE risk, and immune cell infiltration analysis and compound molecular docking studies were performed on the core gene ISG15. Results The study successfully screened 3,456 DEGs and identified core gene modules highly related to SLE through WGCNA analysis, including key genes closely related to the pathogenesis of SLE, such as STAT1, DDX58, ISG15, IRF7, and IFIH1. In particular, this study found a significant positive correlation between the ISG15 gene and SLE, suggesting that it may be a potential risk factor for SLE. Additionally, through molecular docking technology, it was discovered that the ISG15 gene can effectively bind to two compounds, genistein, and flavopiridol, which have anti-inflammatory and immunosuppressive effects, respectively. This provides new potential drug targets for SLE treatment. Discussion As an immunomodulatory cytokine, ISG15 plays a crucial role in the pathogenesis of SLE. This study found that variations in the ISG15 gene may increase the risk of SLE and exacerbate inflammatory responses and tissue damage through multiple mechanisms. Furthermore, molecular docking revealed that genistein and flavopiridol can effectively bind to ISG15, offering a new approach for SLE treatment. These two compounds, with their anti-inflammatory and immunosuppressive properties, have the potential to slow the progression of SLE by influencing the expression and function of ISG15. Conclusion Through comprehensive bioinformatics analysis and Mendelian randomization analysis, this study deeply explored the molecular pathogenic mechanism of SLE and successfully identified ISG15 as a potential therapeutic target for SLE. Simultaneously, molecular docking technology revealed that two compounds, genistein and flavopiridol, have potential therapeutic effects with ISG15, providing new potential drugs for SLE treatment. These discoveries not only enhance our understanding of the pathogenesis of SLE but also provide important clues for developing new treatment strategies.
Collapse
Affiliation(s)
- Aishanjiang Apaer
- Department of Pharmacy, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | - Yanyan Shi
- Department of Pharmacy, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | | | - Fujie Wen
- Department of Pediatrics, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | - Adalaiti Muhetaer
- Department of Pharmacy, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | - Nuermaimaiti Ajimu
- Department of Anesthesiology, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | - Maierhaba Sulitan
- Department of Pediatrics, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| | - Lei Cheng
- Department of Pharmacy, The First People’s Hospital of Kashi Prefecture, Xinjiang, China
| |
Collapse
|
3
|
Zhang Y, Wang J, Fang Y, Liang W, Lei L, Wang J, Gao X, Ma C, Li M, Guo H, Wei L. IFN-α affects Th17/Treg cell balance through c-Maf and associated with the progression of EBV- SLE. Mol Immunol 2024; 171:22-35. [PMID: 38749236 DOI: 10.1016/j.molimm.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease, of which the pathogens is remains obscure. Viral infection, particularly Epstein Barr viru (EBV) infection, has been considered a common pathogenic factor. This study suggests that c-Maf may be an important target in T cell differentiation during SLE progression, providing a potentially new perspective on the role of viral infection in the pathogenesis of autoimmune diseases. METHODS Cytokines of EBV-infected SLE patients were measured by ELISA and assessed in conjunction with their clinical data. IFN-α, c-Maf, and the differentiation of Th17/Treg cells in SLE patients and MRL/LPR mice were analyzed using FCM, WB, RT-PCR, etc. Following the infection of cells and mice with EBV or viral mimic poly (dA:dT), the changes of the aforementioned indicators were investigated. The relationship among IFN-α, STAT3, c-Maf and Th17 cells was determined by si-RNA technique. RESULTS Many SLE patients are found to be complicated by viral infections; Further, studies have demonstrated that viral infection, especially EBV, is involved in SLE development. This study showed that viral infections might promote IFN-α secretion, inhibit c-Maf expression by activating STAT3, increase Th17 cell differentiation, and lead to the immune imbalance of Th17/Treg cells, thus playing a role in the onset and progression of SLE. CONCLUSION This study demonstrates that EBV infections may contribute to SLE development by activating STAT3 through IFN-α, inhibiting c-Maf, and causing Th17/Treg immune imbalance. Our work provided a new insight into the pathogenesis and treatment of SLE.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China; Department of Rheumatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiachao Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Yaqi Fang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Wenzhang Liang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Lingyan Lei
- Department of Rheumatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Junhai Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Xue Gao
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Cuiqing Ma
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Miao Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China
| | - Huifang Guo
- Department of Rheumatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Lin Wei
- Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
4
|
Paul K, Hernández-Armengol R, Lee JY, Chang CY, Shibata T, Yamashita M, Jefferies C, Gibb DR. Distinct RBC alloantibody responses in type 1 interferon-dependent and -independent lupus mouse models. Front Immunol 2024; 14:1304086. [PMID: 38288124 PMCID: PMC10822987 DOI: 10.3389/fimmu.2023.1304086] [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: 10/19/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
During transfusion of red blood cells (RBCs), recipients are exposed to both ABO and non-ABO 'minor' antigens. RBC donor units and recipient RBCs are not routinely matched for non-ABO antigens. Thus, recipients are exposed to many RBC alloantigens that can lead to RBC alloantibody production and subsequent clinically significant hemolysis. RBC alloantibodies also significantly limit the provision of compatible RBC units for recipients. Prior studies indicate that the frequency of RBC alloimmunization is increased during inflammatory responses and in patients with autoimmune diseases. Still, mechanisms contributing to alloimmune responses in patients with autoimmunity are not well understood. More than half of adult patients with systemic lupus erythematosus (SLE) produce type 1 interferons (IFNα/β) and express IFNα/β stimulated genes (ISGs). Previously, we reported that IFNα/β promote RBC alloimmune responses in the pristane mouse model, which develops a lupus-like phenotype that is dependent on IFNα/β signaling. However, it is unclear whether IFNα/β or the lupus-like phenotype induces alloimmunization in lupus models. Therefore, we tested the hypothesis that IFNα/β promotes RBC alloimmune responses in lupus by examining alloimmune responses in IFNα/β-independent (MRL-lpr) and IFNα/β-dependent (pristane) lupus models. Whereas pristane treatment significantly induced interferon-stimulated genes (ISGs), MRL-lpr mice produced significantly lower levels that were comparable to levels in untreated WT mice. Transfusion of murine RBCs that express the KEL antigen led to anti-KEL IgG production by pristane-treated WT mice. However, MRL-lpr mice produced minimal levels of anti-KEL IgG. Treatment of MRL-lpr mice with recombinant IFNα significantly enhanced alloimmunization. Collectively, results indicate that a lupus-like phenotype in pre-clinical models is not sufficient to induce RBC alloantibody production, and IFNα/β gene signatures may be responsible for RBC alloimmune responses in lupus mouse models. If these findings are extended to alternate pre-clinical models and clinical studies, patients with SLE who express an IFNα/β gene signature may have an increased risk of developing RBC alloantibodies and may benefit from more personalized transfusion protocols.
Collapse
Affiliation(s)
- Kausik Paul
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rosario Hernández-Armengol
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - June Young Lee
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Che-Yu Chang
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Tomohiro Shibata
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Michifumi Yamashita
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Caroline Jefferies
- Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David R. Gibb
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
5
|
Arnaud L, Furie R, Morand EF, Aringer M, Peschken C, Desta B, Rapsomaniki E, Hedberg J, Knagenhjelm J, Seo C, Grünfeld Eén T, Sorrentino A, Tummala R, Stirnadel-Farrant HA, Ding B. Burden of systemic lupus erythematosus in clinical practice: baseline data from the SLE Prospective Observational Cohort Study (SPOCS) by interferon gene signature. Lupus Sci Med 2023; 10:e001032. [PMID: 38123459 DOI: 10.1136/lupus-2023-001032] [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: 08/23/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE The longitudinal Systemic Lupus Erythematosus Prospective Observational Cohort Study (SPOCS) aims to assess SLE disease course overall and according to type I interferon 4 gene signature (IFNGS). Here, we describe SPOCS patient characteristics by IFNGS and baseline disease activity. METHODS SPOCS (NCT03189875) is an international study of patients with SLE according to Systemic Lupus International Collaborating Clinics (SLICC)/American College of Rheumatology (ACR) criteria. Enrolled patients from 135 centres in 8 countries were followed biannually for ≤3 years from June 2017 to November 2022. Baseline demographics, disease characteristics, organ system involvement/damage and flares were analysed descriptively according to SLE Disease Activity Index-2000 score (SLEDAI-2K <10/≥10) and IFNGS status (high/low). RESULTS The study population (n=823) was 93.2% female, with mean (SD) age 45.3 (13.9) years and 11.1 (9.2) years since diagnosis; 52.4% had baseline SLICC/ACR Damage Index score ≥1. Patients with SLEDAI-2K scores ≥10 (241 of 584, 41.3%) vs <10 were younger (mean 42.8 (13.7) vs 46.6 (14.2) years; nominal p=0.001), had shorter SLE duration (10.4 (8.6) vs 12.4 (9.6) years; nominal p=0.012) and more severe flares (12.9% vs 5.3%; nominal p=0.001). IFNGS-high patients (522 of 739, 70.6%) were younger than IFNGS-low patients at first SLE manifestation (30.0 (12.7) vs 36.8 (14.6) years; nominal p<0.001). Proportions of IFNGS-high patients differed according to race (nominal p<0.001), with higher proportions among Asian (83.3%) and black (86.5%) versus white patients (63.5%). Greater proportions of IFNGS-high versus IFNGS-low patients had haematological (12.6% vs 4.1%), immunological (74.4% vs 45.6%) or dermal (69.7% vs 62.2%) involvement. CONCLUSIONS We identified key characteristics of patients with high disease activity and/or elevated type I IFN signalling, populations with SLE with high unmet needs. Baseline SLEDAI-2K ≥10 was associated with shorter disease duration and more severe flares. IFNGS-high patients were younger at diagnosis and had distinct patterns of organ involvement, compared with IFNGS-low patients.
Collapse
Affiliation(s)
- Laurent Arnaud
- Department of Rheumatology, University Hospitals of Strasbourg and French National Reference Center for Rare Autoimmune Diseases (RESO), INSERM UMR-S 1109, Strasbourg, France
| | - Richard Furie
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | | | - Martin Aringer
- University Medical Center and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Christine Peschken
- Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Barnabas Desta
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | | | - Caroline Seo
- BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | - Raj Tummala
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Bo Ding
- BioPharmaceuticals Medical, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
6
|
Yang Y, Song J, Zhao H, Zhang H, Guo M. Patients with dermatomyositis shared partially similar transcriptome signature with COVID-19 infection. Autoimmunity 2023; 56:2220984. [PMID: 37353938 DOI: 10.1080/08916934.2023.2220984] [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: 02/08/2023] [Accepted: 05/28/2023] [Indexed: 06/25/2023]
Abstract
Dermatomyositis (DM) is an autoimmune disease that primarily affects the skin and skeletal muscle. Virus infection and type I interferon-related signaling pathways play an important role in the pathogenesis of dermatomyositis. In this study, we found that the skin of patients with DM and the skin of patients with COVID-19 have similar transcriptional profiles, and identified key genes involved in dermatomyositis based on bioinformatics analysis. These hub-genes might be served as potential biomarkers for the early diagnosis and therapy of DM, including MX1, ISG15, IFIT3, IFIT1, RSAD2, IFIT2, IFI6, XAF1, IRF9, MX2.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Jie Song
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Hongjun Zhao
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
7
|
Pan L, Liu J, Liu C, Guo L, Punaro M, Yang S. Childhood-onset systemic lupus erythematosus: characteristics and the prospect of glucocorticoid pulse therapy. Front Immunol 2023; 14:1128754. [PMID: 37638017 PMCID: PMC10448525 DOI: 10.3389/fimmu.2023.1128754] [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: 12/21/2022] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Childhood-onset systemic lupus erythematosus (cSLE) is an autoimmune disease that results in significant damage and often needs more aggressive treatment. Compared to adult-onset SLE, cSLE has a stronger genetic background and more prevalent elevated type I Interferon expression. The management of cSLE is more challenging because the disease itself and treatment can affect physical, psychological and emotional growth and development. High dose oral glucocorticoid (GC) has become the rule for treating moderate to severe cSLE activity. However, GC-related side effects and potential toxicities are problems that cannot be ignored. Recent studies have suggested that GC pulse therapy can achieve disease remission rapidly and reduce GC-related side effects with a reduction in oral prednisone doses. This article reviews characteristics, including pathogenesis and manifestations of cSLE, and summarized the existing evidence on GC therapy, especially on GC pulse therapy in cSLE, followed by our proposal for GC therapy according to the clinical effects and pathogenesis.
Collapse
Affiliation(s)
- Lu Pan
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital, Jilin University, Changchun, China
| | - Jinxiang Liu
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital, Jilin University, Changchun, China
| | - Congcong Liu
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital, Jilin University, Changchun, China
| | - Lishuang Guo
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital, Jilin University, Changchun, China
| | - Marilynn Punaro
- Pediatric Rheumatology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Rheumatology, Texas Scottish Rite Hospital for Children, Houston, TX, United States
- Pediatric Rheumatology, Children’s Medical Center of Dallas, Dallas, TX, United States
| | - Sirui Yang
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
8
|
Novel polymorphism of IFI44L associated with the susceptibility and clinical characteristics of systemic lupus erythematosus in a Chinese population. Int Immunopharmacol 2023; 117:109979. [PMID: 36893516 DOI: 10.1016/j.intimp.2023.109979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Interferon-inducible 44 like (IFI44L) is a newly discovered gene which has been reported to associate with the susceptibility of some infectious diseases, but there is no data on IFI44L SNP polymorphism associated with Systemic lupus erythematosus (SLE). In this study, we aimed to evaluate the association of IFI44L rs273259 polymorphism with the susceptibility and clinical characteristics of SLE in a Chinese population. METHODS 576 SLE patients and 600 controls were recruited in this case-control study. Blood DNA was extracted and IFI44L rs273259 polymorphism was detected by TaqMan SNP Genotyping Assay Kit. The expression levels of IFI44L in Peripheral blood mononuclear cells were detected by RT-qPCR. The DNA methylation levels of IFI44L promoter were detected by bisulfite pyrosequencing. RESULTS The genotype and allele frequencies of IFI44L rs273259 in SLE patients have a significantly difference compared to healthy controls (P < 0.001). The genotype AG (vs. AA: OR = 2.849; P < 0.001) and the allele G (vs. A: OR = 1.454; P < 0.001) were associated with increased susceptibility of SLE. IFI44L rs273259 polymorphism was associated with clinical characteristics of SLE including malar rash (P < 0.001), discoid rash (P < 0.001), lupus nephritis (P < 0.001) and anti-Smith antibodies (P < 0.001). The expression levels of IFI44L were most significantly increased in genotype AG than genotype AA and GG (P < 0.01). The DNA methylation levels of IFI44L promoter were most significantly decreased in genotype AG than genotype AA and GG (P < 0.01). CONCLUSIONS Our results indicate novel polymorphism of IFI44L rs273259 was associated with the susceptibility and clinical characteristics of SLE in the Chinese population.
Collapse
|
9
|
Integrative Bioinformatics Analysis Identifies DDX60 as a Potential Biomarker for Systemic Lupus Erythematosus. DISEASE MARKERS 2023; 2023:8564650. [PMID: 36655136 PMCID: PMC9842429 DOI: 10.1155/2023/8564650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 01/11/2023]
Abstract
Background Systemic lupus erythematosus (SLE) is an autoimmune disease with strong heterogeneity, leading to variable clinical symptoms, which makes diagnosis and activity evaluation difficult. Methods The original dataset of GSE88884 was analyzed to screen differentially expressed genes (DEGs) of SLE and the correlation between DEGs and clinical parameters (SLEDAI, anti-dsDNA, C3, and C4). The result was validated by microarray GSE121239 and SLE patients with RT-qPCR. Next, receiver operator characteristic (ROC) analysis, correlation analysis, and ordinal logistic regression were applied, respectively, to evaluate the capability of diagnosis and prediction of the candidate biomarker. Subsequently, the biological functions of the candidate biomarker were investigated through KEGG and GO enrichment, protein-protein interaction network, and the correlation matrix. Results A total of 283 DEGs were screened, and seven of them were overlapped with SLE-related genes. DDX60 was identified as the candidate biomarker. Analyses of GSE88884, GSE121239, and SLE patients with RT-qPCR indicated that DDX60 expression level is significantly higher in patients with high disease activity. ROC analysis and the area under the ROC curve (AUC = 0.8818) suggested that DDX60 has good diagnostic performance. DDX60 expression level was positively correlated with SLEDAI scores (r = 0.24). For every 1-unit increase in DDX60 expression value, the odds of a higher stage of activity of SLE disease are multiplied by 1.47. The function of DDX60 mainly focuses on IFN-I-induced antiviral activities, RIG-I signaling, and innate immune. Moreover, DDX60 plays a synergistic role with DDX58, IFIH1, OASL, IFIT1, and other related genes in the SLE pathogenesis. Conclusions. DDX60 is differently expressed in SLE, and it is significantly related to both serological indicators and the disease activity of SLE. We suggested that DDX60 might be a potential biomarker for SLE diagnosis and management.
Collapse
|
10
|
Shen M, Duan C, Xie C, Wang H, Li Z, Li B, Wang T. Identification of key interferon-stimulated genes for indicating the condition of patients with systemic lupus erythematosus. Front Immunol 2022; 13:962393. [PMID: 35967341 PMCID: PMC9365928 DOI: 10.3389/fimmu.2022.962393] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with highly heterogeneous clinical symptoms and severity. There is complex pathogenesis of SLE, one of which is IFNs overproduction and downstream IFN-stimulated genes (ISGs) upregulation. Identifying the key ISGs differentially expressed in peripheral blood mononuclear cells (PBMCs) of patients with SLE and healthy people could help to further understand the role of the IFN pathway in SLE and discover potential diagnostic biomarkers.The differentially expressed ISGs (DEISG) in PBMCs of SLE patients and healthy persons were screened from two datasets of the Gene Expression Omnibus (GEO) database. A total of 67 DEISGs, including 6 long noncoding RNAs (lncRNAs) and 61 messenger RNAs (mRNAs) were identified by the “DESeq2” R package. According to Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, those DEISGs were mainly concentrated in the response to virus and immune system processes. Protein-protein interaction (PPI) network showed that most of these DEISGs could interact strongly with each other. Then, IFIT1, RSAD2, IFIT3, USP18, ISG15, OASL, MX1, OAS2, OAS3, and IFI44 were considered to be hub ISGs in SLE by “MCODE” and “Cytohubba” plugins of Cytoscape, Moreover, the results of expression correlation suggested that 3 lncRNAs (NRIR, FAM225A, and LY6E-DT) were closely related to the IFN pathway.The lncRNA NRIR and mRNAs (RSAD2, USP18, IFI44, and ISG15) were selected as candidate ISGs for verification. RT-qPCR results showed that PBMCs from SLE patients had substantially higher expression levels of 5 ISGs compared to healthy controls (HCs). Additionally, statistical analyses revealed that the expression levels of these ISGs were strongly associated to various clinical symptoms, including thrombocytopenia and facial erythema, as well as laboratory indications, including the white blood cell (WBC) count and levels of autoantibodies. The Receiver Operating Characteristic (ROC) curve demonstrated that the IFI44, USP18, RSAD2, and IFN score had good diagnostic capabilities of SLE.According to our study, SLE was associated with ISGs including NRIR, RSAD2, USP18, IFI44, and ISG15, which may contribute to the future diagnosis and new personalized targeted therapies.
Collapse
Affiliation(s)
- Mengjia Shen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Congcong Duan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Changhao Xie
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Hongtao Wang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Zhijun Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Baiqing Li
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Tao Wang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
- *Correspondence: Tao Wang,
| |
Collapse
|
11
|
Huang F, Thokerunga E, He F, Zhu X, Wang Z, Tu J. Research progress of the application of mesenchymal stem cells in chronic inflammatory systemic diseases. Stem Cell Res Ther 2022; 13:1. [PMID: 34998430 PMCID: PMC8742935 DOI: 10.1186/s13287-021-02613-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/04/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic inflammatory systemic diseases are the result of the body's immune imbalance, with a long course and recurring episodes. Immunosuppressants are the main treatment, but not all patients respond well to it. Being capable of both self-renewal and differentiation into multiple tissue cells and low immunogenicity, mesenchymal stem cell is a promising treatment for chronic inflammatory systemic diseases. In this article, we describe the research progress and clinical application of mesenchymal stem cells in chronic inflammatory systemic diseases and look for influencing factors and biomarkers that can predict the outcome of patient with mesenchymal stem cell transplantation.
Collapse
Affiliation(s)
- Fangfang Huang
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Erick Thokerunga
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fajian He
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinyu Zhu
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zi Wang
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiancheng Tu
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
12
|
Differentiating between UCTD and early-stage SLE: from definitions to clinical approach. Nat Rev Rheumatol 2022; 18:9-21. [PMID: 34764455 DOI: 10.1038/s41584-021-00710-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with heterogeneous clinical manifestations that can potentially affect every organ and system. SLE is usually identified on the basis of clinical or serological manifestations; however, some individuals can present with signs and symptoms that are consistent with SLE but are not sufficient for a definite diagnosis. Disease in these individuals can either progress over time to definite SLE or remain stable, in which case their disease is often described as intermediate, possible or probable SLE. Alternatively, such individuals might have undifferentiated connective tissue disease (UCTD). Being able to differentiate between those with stable UCTD and those with SLE at an early stage is important to avoid irreversible target-organ damage from occurring. This Review provides insight into existing and evolving perceptions of the early stages of SLE, including clinical and mechanistic considerations, as well as potential paths towards early identification and intervention. Further research into the earliest phases of SLE will be important for the development of targeted diagnostic approaches and biomarkers for the identification of individuals with early disease who are likely to progress to definite SLE.
Collapse
|
13
|
Jeong S, Lee YS, Kim K, Yoon JS, Kim S, Ha J, Kang I, Choe W. 2-O-Methylhonokiol Suppresses HCV Replication via TRAF6-Mediated NF-kB Activation. Int J Mol Sci 2021; 22:ijms22126499. [PMID: 34204438 PMCID: PMC8234778 DOI: 10.3390/ijms22126499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023] Open
Abstract
Hepatitis C virus (HCV) is associated with various liver diseases. Chronic HCV infection is characterized by an abnormal host immune response. Therefore, it is speculated that to suppress HCV, a well-regulated host immune response is necessary. 2-O-methylhonokiol was identified by the screening of anti-HCV compounds using Renilla luciferase assay in Huh 7.5/Con 1 genotype 1b replicon cells. Here, we investigated the mechanism by which 2-O-methylhonokiol treatment inhibits HCV replication using real-time PCR. Our data shows that treatment with 2-O-methylhonokiol activated innate immune responses via nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) pathway. Additionally, the immunoprecipitation result shows that treatment with 2-O-methylhonokiol augmented tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6) by preventing p62 from binding to TRAF6, resulting in reduced autophagy caused by HCV. Finally, we reproduced our data with the conditioned media from 2-O-methylhonokiol-treated cells. These findings strongly suggest that 2-O-methylhonokiol enhances the host immune response and suppresses HCV replication via TRAF6-mediated NF-kB activation.
Collapse
Affiliation(s)
- Suyun Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
| | - Young-seok Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
| | - Kiyoon Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Ji-su Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
| | - Sungsoo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Joohun Ha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Wonchae Choe
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.J.); (Y.-s.L.); (J.-s.Y.); (S.K.); (J.H.); (I.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-0940
| |
Collapse
|
14
|
Singh RP, Hahn BH, Bischoff DS. Effects of Peptide-Induced Immune Tolerance on Murine Lupus. Front Immunol 2021; 12:662901. [PMID: 34093553 PMCID: PMC8171184 DOI: 10.3389/fimmu.2021.662901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
The regulation of autoimmunity and the molecular mechanisms by which different immune cells, including T cells, polymorphonuclear leukocytes (PMN-granulocytes), and B cells suppress autoimmune diseases is complex. We have shown previously that BWF1 lupus mice are protected from autoimmunity after i.v. injection or oral administration of tolerogenic doses of pCons, an artificial synthetic peptide based on sequences containing MHC class I and MHC class II determinants in the VH region of a J558-encoded BWF1 anti-DNA Ab. Several T cell subsets can transfer this tolerance. In this study, we determined the potential roles of granulocytes, B cells and regulatory T cells altered by pCons treatment in the BWF1 (NZB/NZW) mouse model of lupus. Immunophenotyping studies indicated that pCons treatment of BWF1 mice significantly increased CD4+FoxP3+ T cells, reduced the percent of B cells expressing CD19+CD5+ but increased the percent of CD19+CD1d+ regulatory B cells and increased the ability of the whole B cell population to suppress IgG anti-DNA production in vitro. pCons treatment significantly decreased the expression of CTLA-4 (cytotoxic T-lymphocyte-associated protein-4) in CD8+ T cells. In addition, peptide administration modified granulocytes so they became suppressive. We co-cultured sorted naïve B cells from mice making anti-DNA Ab (supported by addition of sorted naive CD4+ and CD8+ T cells from young auto-antibody-negative BWF1 mice) with sorted B cells or granulocytes from tolerized mice. Both tolerized granulocytes and tolerized B cells significantly suppressed the production of anti-DNA in vitro. In granulocytes from tolerized mice compared to saline-treated littermate controls, real-time PCR analysis indicated that expression of interferon-induced TNFAIP2 increased more than 2-fold while Ptdss2 and GATA1 mRNA were up-regulated more than 10-fold. In contrast, expression of these genes was significantly down-regulated in tolerized B cells. Further, another IFN-induced protein, Bcl2, was reduced in tolerized B cells as determined by Western blot analyses. In contrast, expression of FoxP3 was significantly increased in tolerized B cells. Together, these data suggest that B cells and granulocytes are altered toward suppressive functions by in vivo tolerization of BWF1 mice with pCons and it is possible these cell types participate in the clinical benefits seen in vivo.
Collapse
Affiliation(s)
- Ram P Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bevra H Hahn
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David S Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Wardowska A. m6A RNA Methylation in Systemic Autoimmune Diseases-A New Target for Epigenetic-Based Therapy? Pharmaceuticals (Basel) 2021; 14:ph14030218. [PMID: 33807762 PMCID: PMC8001529 DOI: 10.3390/ph14030218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
The general background of autoimmune diseases is a combination of genetic, epigenetic and environmental factors, that lead to defective immune reactions. This erroneous immune cell activation results in an excessive production of autoantibodies and prolonged inflammation. During recent years epigenetic mechanisms have been extensively studied as potential culprits of autoreactivity. Alike DNA and proteins, also RNA molecules are subjected to an extensive repertoire of chemical modifications. N6-methyladenosine is the most prevalent form of internal mRNA modification in eukaryotic cells and attracts increasing attention due to its contribution to human health and disease. Even though m6A is confirmed as an essential player in immune response, little is known about its role in autoimmunity. Only few data have been published up to date in the field of RNA methylome. Moreover, only selected autoimmune diseases have been studied in respect of m6A role in their pathogenesis. In this review, I attempt to present all available research data regarding m6A alterations in autoimmune disorders and appraise its role as a potential target for epigenetic-based therapies.
Collapse
Affiliation(s)
- Anna Wardowska
- Department of Embryology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
16
|
Zhu J, Xiao J, Wang M, Hu D. Pan-Cancer Molecular Characterization of m 6A Regulators and Immunogenomic Perspective on the Tumor Microenvironment. Front Oncol 2021; 10:618374. [PMID: 33585244 PMCID: PMC7876474 DOI: 10.3389/fonc.2020.618374] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/04/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose N6-methyladenosine (m6A) methylation plays a critical role in diverse biological processes. However, knowledge regarding the constitution of m6A on tumor microenvironment (TME) and tumor-infiltrating lymphocytes (TILs) across cancer types is still lacking. We performed comprehensive immuno-genomic analyses to reveal molecular characterization of the m6A regulators and immune-related genes (IRGs) across TME and TIL heterogeneity. Methods We comprehensively analyzed the properties of m6A regulators in genomic profiles from The Cancer Genome Atlas (TCGA) according to expression perturbations of crucial IRGs, CD274, CD8A, GZMA, and PRF1. The four IRGs were proved to be reliable biomarkers of TILs and TME via CIBERSORT and ESTIMATE analyses, and their co-expression relationship was certified by TIMER analysis. Based on their median values, the samples from the pan-cancer tissues (N = 11,057) were classified into eight TME types. The RNA expression levels of 13 m6A regulators were compared across TME subtypes. Single-sample Gene Set Enrichment Analysis (ssGSEA) was also used to classify TME clusters, expression variants of IRGs and m6A regulators were verified among TME clusters. Meanwhile, the correlation between m6A regulators and tumor mutational burden (TMB) were tested. Finally, the impacts of IRGs and TME clusters in clinical characteristics and outcomes were revealed. Results CD274, CD8A, GZMA, and PRF1 showed similar TILs' characteristics, of which the level of T cells CD8 and T cells CD4 memory activated are consistent with the expression levels of the four IRGs and higher immune infiltration. Besides, CD274, CD8A, GZMA, and PRF1 were positively correlated with the stromal score or immune score in almost all 33 tumor types. All of four IRGs showed impact between tumor pathological stages or clinical outcomes. Among TME type I to type IV, m6A regulators' expression drift changed from high-level to low-level in ESCA, BLCA, HNSC, CESC, BRCA, and GBM. However among TME type V to type VIII, m6A regulators drew a shift from low-level to high-level expression in CESC, BLCA, ESCA, KIRP, HNSC, BRCA, KIRC, COAD, LAML, GBM, and KICH. In ssGSEA analyses, IRGs' expression levels were elevated with the immune infiltration degree and m6A regulators' expression level varied among three TIL subgroups. With different TMB levels, expression differences of m6A regulators were observed in BLCA, BRCA, COAD, LGG, LUAD, LUSC, STAD, THCA, and UCEC. Conclusion We identified four crucial IRGs affecting TILs, TME characteristics and clinical parameters. Expression variants of m6A regulators among the subgroups of TME types and ssGSEA clusters suggested that m6A regulators may be essential factors for phenotypic modifications of IRGs and thus affecting TME characteristics across multiple tumor types.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Intensive Care Unit, The People's Hospital of Tongliang District, Chongqing, China
| | - Jiani Xiao
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Min Wang
- Department of Respiratory and Geriatrics, Chongqing Public Health Medical Center, Chongqing, China
| | - Daixing Hu
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Khan H, Ullah H, Khattak S, Aschner M, Aguilar CN, Halimi SMA, Cauli O, Shah SMM. Therapeutic potential of alkaloids in autoimmune diseases: Promising candidates for clinical trials. Phytother Res 2021; 35:50-62. [PMID: 32667693 DOI: 10.1002/ptr.6763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/23/2020] [Accepted: 05/16/2020] [Indexed: 02/05/2023]
Abstract
Clinical investigations have characterized numerous disorders like autoimmune diseases, affecting the population at a rate of approximately 8-10%. These disorders are characterized by T-cell and auto-antibodies responses to self-molecules by immune system reactivity. Several therapeutic options have been adopted in clinics to combat such diseases, however, most of them are recurring. Thus, the discovery of new effective agents for the treatment of autoimmune diseases is paramount. In this context, natural products might be a useful alternative to the current therapies. Plant alkaloids with their substantial therapeutic history can be particularly interesting candidates for the alleviation of autoimmune ailments. This review encompasses various alkaloids with significant effects against autoimmune diseases in preclinical trials. These results suggest further clinical assessment with respect to autoimmune illnesses. Furthermore, the application of modern technologies such as nanoformulation could be also helpful in the design of more effective therapies and thus further studies are needed to decipher their therapeutic efficacy as well as potential limitations.
Collapse
Affiliation(s)
- Haroon Khan
- Abdul Wali khan university Mardan, Abdul Wali khan university Mardan, Department of Pharmacy, Abdul Wali Khan university Mardan, Pakistan, Mardan, Pakistan, 23200, Pakistan
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Sumaira Khattak
- Abdul Wali khan university Mardan, Abdul Wali khan university Mardan, Department of Pharmacy, Abdul Wali Khan university Mardan, Pakistan, Mardan, Pakistan, 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Cristobal N Aguilar
- School of Chemistry, Universidad Autónoma de Coahuila Saltillo, Saltillo, Mexico
| | - Syed M A Halimi
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Omar Cauli
- Department of Nursing, University of Valencia, Valencia, Spain
| | - Syed M M Shah
- Department of Pharmacy, University of Swabi Pakistan, Swabi, Pakistan
| |
Collapse
|
18
|
Li J, Li S, Yang L, Cao P, Lu J. Severe fever with thrombocytopenia syndrome virus: a highly lethal bunyavirus. Crit Rev Microbiol 2020; 47:112-125. [PMID: 33245676 DOI: 10.1080/1040841x.2020.1847037] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel bunyavirus. Since 2007, SFTS disease has been reported in China with high fatality rate up to 30%, which drew high attention from Centre for Disease Control and Prevention and government. SFTSV is endemic in the centra l and eastern China, Korea and Japan. There also have been similar cases reported in Vietnam. The number of SFTSV infection cases has a steady growth in these years. As SFTSV could transmitted from person to person, it will expose the public to infectious risk. In 2018 annual review of the Blueprint list of priority diseases, World Health Organisation has listed SFTSV infection as prioritised diseases for research and development in emergency contexts. However, the pathogenesis of SFTSV remains largely unclear. Currently, there are no specific therapeutics or vaccines to combat infections of SFTSV. This review discusses recent findings of epidemiology, transmission pathway, pathogenesis and treatments of SFTS disease.
Collapse
Affiliation(s)
- Jing Li
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Shen Li
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Li Yang
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Pengfei Cao
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jianhong Lu
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
19
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Hammond ER, Tummala R, Berglind A, Syed F, Wang X, Desta B, Nab H. Study protocol for the international Systemic Lupus Erythematosus Prospective Observational Cohort Study (SPOCS): understanding lupus and the role of type I interferon gene signature. BMJ Open 2020; 10:e036563. [PMID: 32873668 PMCID: PMC7467530 DOI: 10.1136/bmjopen-2019-036563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/23/2020] [Accepted: 06/03/2020] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The Systemic Lupus Erythematosus (SLE) Prospective Observational Cohort Study (SPOCS) aims to describe the disease course of SLE and its association with type I interferon gene signature (IFNGS) status. METHODS AND ANALYSIS SPOCS is an international, multicentre, prospective, observational cohort study designed to follow patients through biannual study visits during a 3-year observation period. Patients ≥18 years old with a physician diagnosis that meets the American College of Rheumatology or Systemic Lupus International Collaborating Clinics SLE classification criteria will be included. SPOCS will comprehensively analyse clinical features, disease progression and treatment, SLE outcomes, health status assessments and quality of life, and healthcare resource utilisation of patients with moderate to severe SLE. A four-gene test will be used to measure IFNGS status; scores will be compared with a pre-established cut-off. Patients will be stratified by low or high IFNGS expression levels. Enrolment began in June 2017, and study completion is expected in 2022. The total number of anticipated patients was initially planned for 1500 patients and was amended to 900 patients owing to slow accrual of eligible patients. ETHICS AND DISSEMINATION The ethics committee/institutional review board/independent ethics committee at each study site approved the SPOCS protocol prior to study initiation (protocol number: D3461R00001, version 3.0, 26 June 2019). Study findings will be disseminated through peer-reviewed publications and presentations at scientific meetings. TRIAL REGISTRATION NUMBER NCT03189875.
Collapse
Affiliation(s)
- Edward R Hammond
- BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, Maryland, USA
| | - Raj Tummala
- Inflammation, Autoimmunity and Neuroscience, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anna Berglind
- BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Farhat Syed
- Precision Medicine, R&D, AstraZeneca, Cambridge, UK
| | - Xia Wang
- Data Science and AI, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Barnabas Desta
- Global Pricing and Market Access, AstraZeneca, Gaithersburg, Maryland, USA
| | - Henk Nab
- Inflammation and Autoimmunity, AstraZeneca, Cambridge, UK
| |
Collapse
|
21
|
Tocut M, Shoenfeld Y, Zandman-Goddard G. Systemic lupus erythematosus: an expert insight into emerging therapy agents in preclinical and early clinical development. Expert Opin Investig Drugs 2020; 29:1151-1162. [PMID: 32755494 DOI: 10.1080/13543784.2020.1807004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic disease that is potentially fatal. There is no cure for SLE and the medications used are associated with toxic side effects. In the era of revolutionary emerging novel biologic agents, the design and investigation of targeted therapy for these patients is necessary. Novel therapies under investigation in phase II-III clinical trials showed promising results. Therapies can target various pathways involved in SLE including cytokines, signal transduction inhibitors, B-cell depletion and interference with co-stimulation. Of interest is the proof of concept of sequential therapy. AREAS COVERED We performed an extensive literature search via PubMed, Medline, Elsevier Science and Springer Link databases between the years 2014-2020 using the following terms: SLE, novel treatments. We have reviewed 232 articles and selected those articles that (i) focus on phase II-III emerging therapies and (ii) offer new findings from existing therapies, which reveal breakthrough concepts in SLE treatment. EXPERT OPINION It is still difficult to crack the puzzle of a successful SLE treatment approach. New strategies with potential may encompass the targeting of more than one protein. Another way forward is to identify each SLE patient and personalize therapy by clinical manifestations, disease activity, serology and activated protein.
Collapse
Affiliation(s)
- Milena Tocut
- Department of Internal Medicine C, Wolfson Medical Center , Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel
| | - Yehuda Shoenfeld
- Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel.,Center for Autoimmune Diseases, Sheba Medical Center , Ramat Gan, Israel.,I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
| | - Gisele Zandman-Goddard
- Department of Internal Medicine C, Wolfson Medical Center , Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel
| |
Collapse
|
22
|
Xu W, Zhang Y, Wang X, Liu P, Gao D, Gu B, Zhang J, Li C, Ren Q, Yang L, Yuan H, Shen M, Chen X. Clinical features and expression of type I interferon-inducible genes in systemic lupus erythematosus patients harboring rs1143679 polymorphism in China: a single-center, retrospective study. Clin Rheumatol 2020; 40:1093-1101. [PMID: 32785810 DOI: 10.1007/s10067-020-05330-x] [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: 10/09/2019] [Revised: 07/02/2020] [Accepted: 08/04/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVE This case-control study aimed to analyze the clinical features and determine the expression of type I interferon-induced genes in systemic lupus erythematosus (SLE) patients harboring the CD11b rs1143679 single-nucleotide polymorphism (SNP) and elucidate whether it is involved in the relapses of SLE. METHODS One hundred twenty-five relatively inactive SLE patients with SLEDAI scores < 6, including 102 CD11b rs1143679 G allele patients as controls and 23 rs1143679 A allele carriers as cases, were enrolled from the SLE patient specimen bank in the Department of Rheumatology and Immunology. The sample set was retrospectively analyzed for differences in clinical features, and quantitative PCR and Western blot analyses were performed to evaluate the relative expression of type I interferon (IFN)-inducible genes. RESULTS The 24-h urinary protein levels in the case group were significantly elevated, and serum C3 levels were significantly reduced compared with those in the control group (P = 0.019 and P = 0.021, respectively). The relative mRNA levels of IFN-inducible genes IFIT1, IFIT4, and ISG15 in the case group were higher than that in the control group (P = 0.0257, 0.0344, and 0.0311, respectively) and matched with the Western blot results. CONCLUSIONS The relative expression of type I IFN-inducible genes in inactive SLE patients harboring the CD11b rs1143679 polymorphism was higher than that in other lupus patients. These findings suggest that the rs1143679 SNP can precipitate relapses in inactive SLE patients. KEY POINTS • The rs1143679 GA genotype was associated with SLE clinical features. • The rs1143679 GA genotype showed higher interferon-inducible gene expression relative to the GG genotype.
Collapse
Affiliation(s)
- Wenyu Xu
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Yueyue Zhang
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Xiaoqin Wang
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Peiyu Liu
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, China
| | - Dayu Gao
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Bingjie Gu
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Junyu Zhang
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Chunmei Li
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Qijie Ren
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Leilei Yang
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Hai Yuan
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Minning Shen
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China
| | - Xingguo Chen
- Department of Rheumatology and Immunology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, China.
| |
Collapse
|
23
|
Mora-Arias T, Amezcua-Guerra LM. Type III Interferons (Lambda Interferons) in Rheumatic Autoimmune Diseases. Arch Immunol Ther Exp (Warsz) 2020; 68:1. [PMID: 31915933 DOI: 10.1007/s00005-019-00564-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
The last 2 decades have witnessed the discovery and characterization of a new family of cytokines with immunological characteristics similar to those described for type I interferons, type III or lambda interferons. Unraveling the molecular mechanisms underlying each type of interferon has allowed us to understand how some autoimmune diseases can be considered as interferonopathies. Under normal conditions, type III interferons play a key role in the defense against viruses by modulating the functioning of several types of innate and adaptive immune cells. These effects include upregulation of major histocompatibility complex molecules by myeloid dendritic cells, increased functioning of pattern recognition receptors by plasmacytoid dendritic cells, decreased activity of regulatory T cells, enhanced production of antibodies by plasmatic cells and increased expression of chemokines and adhesion molecules by leukocytes and endothelial cells. Notably, all these mechanisms have been described to boost autoimmunity, and type III interferons pathway activation has been related to the pathogenesis of autoimmune conditions such as systemic lupus erythematosus, systemic sclerosis and Sjögren's syndrome. This review provides an overview of the current evidence on the contribution of type III interferons in the pathogenesis of rheumatic autoimmune diseases in humans.
Collapse
Affiliation(s)
- Tania Mora-Arias
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Luis M Amezcua-Guerra
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, 14080, Mexico City, Mexico.
| |
Collapse
|
24
|
Smith N, Rodero MP, Bekaddour N, Bondet V, Ruiz-Blanco YB, Harms M, Mayer B, Bader-Meunier B, Quartier P, Bodemer C, Baudouin V, Dieudonné Y, Kirchhoff F, Sanchez Garcia E, Charbit B, Leboulanger N, Jahrsdörfer B, Richard Y, Korganow AS, Münch J, Nisole S, Duffy D, Herbeuval JP. Control of TLR7-mediated type I IFN signaling in pDCs through CXCR4 engagement-A new target for lupus treatment. SCIENCE ADVANCES 2019; 5:eaav9019. [PMID: 31309143 PMCID: PMC6620093 DOI: 10.1126/sciadv.aav9019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/06/2019] [Indexed: 06/10/2023]
Abstract
Type I interferons are highly potent cytokines essential for self-protection against tumors and infections. Deregulations of type I interferon signaling are associated with multiple diseases that require novel therapeutic options. Here, we identified the small molecule, IT1t, a previously described CXCR4 ligand, as a highly potent inhibitor of Toll-like receptor 7 (TLR7)-mediated inflammation. IT1t inhibits chemical (R848) and natural (HIV) TLR7-mediated inflammation in purified human plasmacytoid dendritic cells from blood and human tonsils. In a TLR7-dependent lupus-like model, in vivo treatment of mice with IT1t drives drastic reduction of both systemic inflammation and anti-double-stranded DNA autoantibodies and prevents glomerulonephritis. Furthermore, IT1t controls inflammation, including interferon α secretion, in resting and stimulated cells from patients with systemic lupus erythematosus. Our findings highlight a groundbreaking immunoregulatory property of CXCR4 signaling that opens new therapeutic perspectives in inflammatory settings and autoimmune diseases.
Collapse
Affiliation(s)
- Nikaïa Smith
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Mathieu P. Rodero
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nassima Bekaddour
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Bondet
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Yasser B. Ruiz-Blanco
- Computational Biochemistry and Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Brigitte Bader-Meunier
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Universitaire Necker, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM UMR 1163, Laboratory of Immunogenetics of Paediatric Autoimmunity, Paris, France
| | - Pierre Quartier
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Universitaire Necker, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christine Bodemer
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Department of Paediatric Dermatology, Reference Centre for Rare Skin Disorders (MAGEC), Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Véronique Baudouin
- Hôpital Universitaire Robert Debré, Néphrologie pédiatrique, Paris, France
| | - Yannick Dieudonné
- CNRS UPR 3572 “Immunopathology and Therapeutic Chemistry”/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
- UFR Medicine, University of Strasbourg, Strasbourg, France
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Elsa Sanchez Garcia
- Computational Biochemistry and Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Bruno Charbit
- Centre for Translational Research, Institut Pasteur, Paris, France
| | - Nicolas Leboulanger
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Pediatric Otolaryngology Department, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Bernd Jahrsdörfer
- Institute of Transfusion Medicine and Immunogenetics (IKT) Ulm, Helmholtzstr. 10, 89081 Ulm, Germany
| | - Yolande Richard
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
| | - Anne-Sophie Korganow
- CNRS UPR 3572 “Immunopathology and Therapeutic Chemistry”/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
- UFR Medicine, University of Strasbourg, Strasbourg, France
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Sébastien Nisole
- IRIM, Université de Montpellier, CNRS UMR, 9004 Montpellier, France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
- Centre for Translational Research, Institut Pasteur, Paris, France
| | - Jean-Philippe Herbeuval
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
25
|
Zhang C, Fu J, Zhou Y. A Review in Research Progress Concerning m6A Methylation and Immunoregulation. Front Immunol 2019; 10:922. [PMID: 31080453 PMCID: PMC6497756 DOI: 10.3389/fimmu.2019.00922] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/10/2019] [Indexed: 12/30/2022] Open
Abstract
Over 100 types of cellular RNA modifications have been identified in both coding and a variety of non-coding RNAs. N6-methyladenosine (m6A) is the most prevalent and abundant post-transcriptional RNA modification on eukaryote mRNA, and its biological functions are mediated by special binding proteins (i.e., methyltransferases, demethylases, and effectors) that recognize this modification. The presence of m6A on transcripts contributes to diverse fundamental cellular functions, such as pre-mRNA splicing, nuclear transport, stability, translation, and microRNA biogenesis, implying an association with numerous human diseases. This review principally summarizes recent progress in the study of m6A methylation mechanisms and relevant roles they play in immunoregulation.
Collapse
Affiliation(s)
- Caiyan Zhang
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinrong Fu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yufeng Zhou
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Association between Type I interferon and depletion and dysfunction of endothelial progenitor cells in C57BL/6 mice deficient in both apolipoprotein E and Fas ligand. Curr Res Transl Med 2018; 66:71-82. [DOI: 10.1016/j.retram.2018.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/31/2017] [Accepted: 02/07/2018] [Indexed: 11/22/2022]
|
27
|
Higher activation of the interferon-gamma signaling pathway in systemic lupus erythematosus patients with a high type I IFN score: relation to disease activity. Clin Rheumatol 2018; 37:2675-2684. [DOI: 10.1007/s10067-018-4138-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/11/2018] [Accepted: 04/20/2018] [Indexed: 12/21/2022]
|
28
|
Wen L, Zhu C, Zhu Z, Yang C, Zheng X, Liu L, Zuo X, Sheng Y, Tang H, Liang B, Zhou Y, Li P, Zhu J, Ding Y, Chen G, Gao J, Tang L, Cheng Y, Sun J, Elango T, Kafle A, Yu R, Xue K, Zhang Y, Li F, Li Z, Guo J, Zhang X, Zhou C, Tang Y, Shen N, Wang M, Yu X, Liu S, Fan X, Gao M, Xiao F, Wang P, Wang Z, Zhang A, Zhou F, Sun L, Yang S, Xu J, Yin X, Cui Y, Zhang X. Exome-wide association study identifies four novel loci for systemic lupus erythematosus in Han Chinese population. Ann Rheum Dis 2018; 77:417. [PMID: 29233832 DOI: 10.1136/annrheumdis-2017-211823] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 11/11/2017] [Accepted: 11/19/2017] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of considerable genetic predisposition. Genome-wide association studies have identified tens of common variants for SLE. However, the majority of them reside in non-coding sequences. The contributions of coding variants have not yet been systematically evaluated. METHODS We performed a large-scale exome-wide study in 5004 SLE cases and 8179 healthy controls in a Han Chinese population using a custom exome array, and then genotyped 32 variants with suggestive evidence in an independent cohort of 13 246 samples. We further explored the regulatory effect of one novel non-coding single nucleotide polymorphism (SNP) in ex vivo experiments. RESULTS We discovered four novel SLE gene regions (LCT, TPCN2, AHNAK2 and TNFRSF13B) encompassing three novel missense variants (XP_016859577.1:p.Asn1639Ser, XP_016859577.1:p.Val219Phe and XP_005267356.1:p.Thr4664Ala) and two non-coding variants (rs10750836 and rs4792801) with genome-wide significance (pmeta <5.00×10-8). These variants are enriched in several chromatin states of primary B cells. The novel intergenic variant rs10750836 exhibited an expression quantitative trait locus effect on the TPCN2 gene in immune cells. Clones containing this novel SNP exhibited gene promoter activity for TPCN2 (P=1.38×10-3) whose expression level was reduced significantly in patients with SLE (P<2.53×10-2) and was suggested to be further modulated by rs10750836 in CD19+ B cells (P=7.57×10-5) in ex vivo experiments. CONCLUSIONS This study identified three novel coding variants and four new susceptibility gene regions for SLE. The results provide insights into the biological mechanism of SLE.
Collapse
Affiliation(s)
- Leilei Wen
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Caihong Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Zhengwei Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Chao Yang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaodong Zheng
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lu Liu
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xianbo Zuo
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yujun Sheng
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Huayang Tang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Bo Liang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yi Zhou
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Pan Li
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jun Zhu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yantao Ding
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Gang Chen
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinping Gao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lili Tang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yuyan Cheng
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jingying Sun
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Tamilselvi Elango
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Anjana Kafle
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Ruixing Yu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Ke Xue
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Yaohua Zhang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Feng Li
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangdong, China
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangdong, China
| | - Shengxiu Liu
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xing Fan
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Min Gao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Fengli Xiao
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Peiguang Wang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Zaixing Wang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Anping Zhang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Fusheng Zhou
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Liangdan Sun
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Sen Yang
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhua Xu
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Xianyong Yin
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Xuejun Zhang
- Department of Dermatology, Institute of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
- Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
- Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
29
|
Thanadetsuntorn C, Ngamjanyaporn P, Setthaudom C, Hodge K, Saengpiya N, Pisitkun P. The model of circulating immune complexes and interleukin-6 improves the prediction of disease activity in systemic lupus erythematosus. Sci Rep 2018; 8:2620. [PMID: 29422675 PMCID: PMC5805742 DOI: 10.1038/s41598-018-20947-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/26/2018] [Indexed: 11/09/2022] Open
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease resulting in autoantibody production, immune complex deposition, and complement activation. The standard biomarkers such as anti-dsDNA and complements (C3 and C4) do not always correlate with active clinical SLE. The heterogeneity of SLE patients may require additional biomarkers to designate disease activity. Ninety SLE patients participated in this study. Evaluation of disease activity was achieved with the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) and modified SLEDAI-2K. The measured serum biomarkers were anti-dsDNA, C3, C4, ESR, interleukin-6 (IL-6), and circulating immune complexes (CIC). IL-6, ESR and CIC significantly increased in active clinical SLE. Complement, anti-dsDNA, ESR and CIC correlated with SLEDAI-2K while only anti-dsDNA, CIC, ESR and IL-6 correlated with modified SLEDAI-2K. A combination of biomarkers gave a higher odds ratio (OR) than any single biomarker. A combination of IL-6 or CIC exhibited the highest OR (OR = 7.27, 95%CI (1.99-26.63), p = 0.003) while either complement or anti-dsDNA showed a moderate odds ratio (OR = 3.14, 95%CI (1.16-8.48), p = 0.024) of predicting clinical active SLE. The combination of CIC and IL-6 strongly predicts active clinical SLE. CIC and IL-6 can be used in addition to standard biomarkers to determine SLE activity.
Collapse
Affiliation(s)
- Chokchai Thanadetsuntorn
- Division of Allergy Immunology and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Pintip Ngamjanyaporn
- Division of Allergy Immunology and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Chavachol Setthaudom
- Immunology Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Kenneth Hodge
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nisara Saengpiya
- Immunology Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy Immunology and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand.
| |
Collapse
|
30
|
Xiao ZX, Zheng X, Hu L, Wang J, Olsen N, Zheng SG. Immunosuppressive Effect of B7-H4 Pathway in a Murine Systemic Lupus Erythematosus Model. Front Immunol 2017; 8:1765. [PMID: 29321778 PMCID: PMC5732181 DOI: 10.3389/fimmu.2017.01765] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
B7-H4, one of the co-stimulatory molecules of the B7 family, has been shown to play an important role in negatively regulating the adaptive immune response by inhibiting the proliferation, activation, and cytokine production of T cells. In this study, we investigate the role of B7-H4 in development of systemic lupus erythematosus (SLE). We investigated a murine model of SLE using transfer of bone marrow-derived dendritic cells (BMDCs) that were incubated with activated syngeneic lymphocyte-derived DNA. The recipient mouse produced anti-ds-DNA antibodies as well as displayed splenomegaly and lymphadenopathy as shown by significantly increased weights, and the kidneys showed lupus-like pathological changes include urine protein and glomerulonephritis with hyperplasia in glomeruli and increased mesangial cells and vasculitis with perivascular cell infiltration, glomerular deposition of IgG and complement C3. We showed that B7-H4 deficiency in BMDCs could cause greater production of anti-ds-DNA antibodies in transferred mice, and the lymph tissue swelling and the kidney lesions were also exacerbated with B7-H4 deficiency. Treatment with a B7-H4 antagonist antibody also aggravated the lupus model. Conversely, B7-H4 Ig alleviated the lupus manifestations. Therefore, we conclude that B7-H4 is a negative check point for the development of SLE in this murine model. These results suggest that this approach may have a clinical potential in treating human SLE.
Collapse
Affiliation(s)
- Ze Xiu Xiao
- Department of Clinical Immunology, Third Hospital at Sun Yat-sen University, Guangzhou, Guangdong, China.,Laboratory of Immunotherapy, Sun Yat-Sen University, Guangzhou, China
| | - Xu Zheng
- Laboratory of Immunotherapy, Sun Yat-Sen University, Guangzhou, China
| | - Li Hu
- Laboratory of Immunotherapy, Sun Yat-Sen University, Guangzhou, China
| | - Julie Wang
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Nancy Olsen
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Song Guo Zheng
- Department of Clinical Immunology, Third Hospital at Sun Yat-sen University, Guangzhou, Guangdong, China.,Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| |
Collapse
|
31
|
Li LJ, Fan YG, Leng RX, Pan HF, Ye DQ. Potential link between m 6A modification and systemic lupus erythematosus. Mol Immunol 2017; 93:55-63. [PMID: 29145159 DOI: 10.1016/j.molimm.2017.11.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 12/14/2022]
Abstract
The field of m6A modification and epitranscriptomics has recently attracted much attention. More methods allowing for precise m6A site profiling and location are developed and crucial players of m6A modification machinery are increasingly identified. Although some challenges remain, m6A modification is found to modulate almost all aspects of RNA metabolism, such as splicing, stability, structure, translation, and export. Thus, m6A modification adds a new layer of post-transcriptional gene expression regulation, and it is implicated in T cell response to HIV infection, type I interferon production, and T cell differentiation and homeostasis. Moreover, evidence supporting its involvement in various human diseases including cancers is accumulating. Given the role of m6A modification in gene expression regulation and immune response, it invites the speculation that m6A modification may justify the pathogenesis of systemic lupus erythematosus (SLE) and take part in the initiation and progression of SLE. In this review, we introduce the widespread existence of m6A modification and briefly discuss components of m6A modification machinery in mammals. We mainly summarize the studies reporting the mechanisms of m6A modification in gene expression regulation through modulating pre-mRNA splicing, mRNA stability, RNA structure, translation, and pri-miRNA processing. Biological functions related to immune response of m6A modification and the implication of m6A modification in cancers are highlighted. In the end, we surmise the potential link between m6A modification and SLE.
Collapse
Affiliation(s)
- Lian-Ju Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China.
| |
Collapse
|
32
|
Wang H, Wang J, Xia Y. Defective Suppressor of Cytokine Signaling 1 Signaling Contributes to the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2017; 8:1292. [PMID: 29085365 PMCID: PMC5650678 DOI: 10.3389/fimmu.2017.01292] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/26/2017] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving injuries in multiple organs and systems. Exaggerated inflammatory responses are characterized as end-organ damage in patients with SLE. Although the explicit pathogenesis of SLE remains unclear, increasing evidence suggests that dysregulation of cytokine signals contributes to the progression of SLE through the Janus kinase/signal transducer and activator of transcription (STAT) signaling pathway. Activated STAT proteins translocate to the cell nucleus and induce transcription of target genes, which regulate downstream cytokine production and inflammatory cell infiltration. The suppressor of cytokine signaling 1 (SOCS1) is considered as a classical inhibitor of cytokine signaling. Recent studies have demonstrated that SOCS1 expression is decreased in patients with SLE and in murine lupus models, and this negatively correlates with the magnitude of inflammation. Dysregulation of SOCS1 signals participates in various pathological processes of SLE such as hematologic abnormalities and autoantibody generation. Lupus nephritis is one of the most serious complications of SLE, and it correlates with suppressed SOCS1 signals in renal tissues. Moreover, SOCS1 insufficiency affects the function of several other organs, including skin, central nervous system, liver, and lungs. Therefore, SOCS1 aberrancy contributes to the development of both systemic and local inflammation in SLE patients. In this review, we discuss recent studies regarding the roles of SOCS1 in the pathogenesis of SLE and its therapeutic implications.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiaxing Wang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Shi X, Jiao B, Chen Y, Li S, Chen L. MxA is a positive regulator of type I IFN signaling in HCV infection. J Med Virol 2017; 89:2173-2180. [PMID: 28561372 DOI: 10.1002/jmv.24867] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 05/11/2017] [Indexed: 01/17/2023]
Abstract
Type I interferons (IFNs) are a family of primordial cytokines that respond to various pathogen infections including Hepatitis C virus (HCV). Type I IFNs signal through Jak/STAT pathway leading to the production of a few hundred interferon stimulated genes (ISGs). The aim of this study was to explore the role of one of these ISGs, MxA in HCV infection and type I IFN production. Plasmid encoding MxA was cloned into PcDNA3.1-3×tag vector and MxA expression was confirmed both at mRNA (RT-PCR) and protein (Western blot, WB) levels. IFNα and IFNβ productions were quantified by RT-PCR from cell lysate and by ELISA kit from culture medium following MxA over-expression in Huh7.5.1 cells. The activation status of Jak/STAT signaling pathway was examined at three levels: p-STAT1 (WB), interferon sensitive response element (ISRE) activity (dual luciferase reporter gene assay), and levels of ISG expression (RT-qPCR). J6/JFH1 HCV culture system was used to study the role of MxA in HCV replication. Our findings indicated that MxA over-expression inhibited HCV replication and potentiated the IFNα-mediated anti-HCV activity; MxA stimulated the production of IFNα, IFNβ, and enhanced IFNα-induced activation of Jak-STAT signaling pathway. We concluded that MxA is a positive regulator of type I IFN signaling in HCV infection.
Collapse
Affiliation(s)
- Xuezhen Shi
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Baihai Jiao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Yanzhao Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Shilin Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Limin Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China.,Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
miRNA-451a Targets IFN Regulatory Factor 8 for the Progression of Systemic Lupus Erythematosus. Inflammation 2017; 40:676-687. [PMID: 28120198 DOI: 10.1007/s10753-017-0514-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increasing evidence has shown that miRNA-451a (miR-451a) is associated with the development of systemic lupus erythematosus (SLE); however, the mechanism of this association is not fully known. The present study found an increased expression of miR-451a in the spleen and thymus of an SLE mice model. A decrease in miR-451a expression partly relieved the enlargement of the spleen and decreased the proteinuria content and immune complex deposits. The deficiency in miR-451a also decreased numbers of CD4+CD69+ and CD4+/CD8+ T cells and the levels of the serum cytokines IL-17a and IL-4. The IFN regulatory factor (IRF) 8 was highly expressed in the immune organs of wild-type mice but was suppressed in SLE-like mice. A dual-luciferase reporter assay was carried out in combination with gene silencing and overexpression to verify that IRF8 was a target of miR-451a in vitro and in vivo. The data indicate the function and a target of miR-451a in SLE, providing a new target for SLE therapy.
Collapse
|