1
|
Ren Y, Chen W, Lin Y, Wang Z, Wang W. Identification of glucocorticoid-related genes in systemic lupus erythematosus using bioinformatics analysis and machine learning. PLoS One 2025; 20:e0319737. [PMID: 40131879 PMCID: PMC11936220 DOI: 10.1371/journal.pone.0319737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/06/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a complex autoimmune disease that has significant impacts on patients' quality of life and poses a substantial economic burden on society. OBJECTIVE This study aimed to elucidate the molecular mechanisms underlying SLE by analyzing glucocorticoid-related genes (GRGs) expression profiles. METHODS We examined the expression profiles of GRGs in SLE and performed consensus clustering analysis to identify stable patient clusters. We also identified differentially expressed genes (DEGs) within the clusters and between SLE patients and healthy controls. We conducted Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) to investigate biological functional differences, and we also conducted CIBERSORTx to estimate the number of immune cells. Furthermore, we utilized least absolute shrinkage and selection operator (LASSO) regression and Random Forest (RF) algorithms to screen for hub genes. We then validated the expression of these hub genes and constructed nomograms for further validation. Moreover, we employed single-sample Gene Set Enrichment Analysis (ssGSEA) to analyze immune infiltration. We also constructed an RNA-binding protein (RBP)-mRNA network and conducted drug sensitivity analysis along with molecular docking studies. RESULTS Patients with SLE were divided into two subclusters, revealing a total of 2,681 DEGs. Among these, 1,458 genes were upregulated, while 1,223 were downregulated in cluster_1. GSVA showed significant changes in the pathways associated with cluster_1. Immune infiltration analysis revealed high levels of monocyte in all samples, with greater infiltration of various immune cells in cluster_1. A comparison of SLE patients to control subjects identified 269 DEGs, which were enriched in several pathways. Hub genes, including PTX3, DYSF and F2R, were selected through LASSO and RF methods, resulting in a well-performing diagnostic model. Drug sensitivity and docking studies suggested F2R as a potential new therapeutic target. CONCLUSION PTX3, DYSF and F2R are potentially linked to SLE and are proposed as new molecular markers for its onset and progression. Additionally, monocyte infiltration plays a crucial role in advancing SLE.
Collapse
Affiliation(s)
- Yinghao Ren
- Department of Dermatology, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China
| | - Weiqiang Chen
- Department of Nephrology, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China
| | - Yuhao Lin
- Department of Endocrinology, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China
| | - Zeyu Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Weiliang Wang
- Epilepsy Center, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China
| |
Collapse
|
2
|
Saito T, Takatsuji R, Murayama G, Yamaji Y, Hagiwara Y, Nishioka Y, Kuga T, Miyashita T, Kusaoi M, Tamura N, Yamaji K. Double-filtration plasmapheresis reduces type I interferon bioavailability and inducing activity in systemic lupus erythematosus. Immunol Med 2024; 47:264-274. [PMID: 38952099 DOI: 10.1080/25785826.2024.2372918] [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: 04/22/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024] Open
Abstract
Type I interferons (IFN-Is) play a significant role in systemic lupus erythematosus (SLE) pathogenesis. Double-filtration plasmapheresis (DFPP) is a treatment option for SLE; however, its effect on IFN-Is remains unclear. Therefore, we investigated the effects of DFPP on IFN-Is. Plasma from patients with SLE (n = 11) who regularly underwent DFPP was analysed using a cell-based reporter system to detect the bioavailability and inducing activity of IFN-I. The concentration of plasma dsDNA was measured, and western blotting analysis was used to assess the phosphorylation of the STING pathway. A higher IFN-I bioavailability and inducing activity were observed in patients compared to healthy controls, and both parameters decreased after DFPP. The reduction in IFN-I-inducing activity was particularly prominent in patients with high disease activity. Notably, this reduction was not observed in STING-knockout reporter cells. Additionally, plasma dsDNA levels decreased after DFPP treatment, suggesting that inhibition of the STING pathway was responsible for the observed decrease in activity. Western blotting analysis revealed suppression of STING pathway phosphorylation after DFPP. DFPP reduced IFN-I bioavailability and the inducing activity of plasma. This reduction is likely attributable to the inhibition of the STING pathway through the elimination of dsDNA.
Collapse
Affiliation(s)
- Takumi Saito
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryo Takatsuji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Goh Murayama
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Yu Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yukitomo Hagiwara
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yujin Nishioka
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Taiga Kuga
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoko Miyashita
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Bulusu SN, Mariaselvam CM, Shah S, Kommoju V, Kavadichanda C, Harichandrakumar KT, Thabah M, Negi VS. Type I interferon gene expression signature as a marker to predict response to cyclophosphamide based treatment in proliferative lupus nephritis. Lupus 2024; 33:1069-1081. [PMID: 39033304 DOI: 10.1177/09612033241266779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
OBJECTIVES To assess the longitudinal effect of cyclophosphamide (CYC) treatment on type-I interferon (IFN) signature in proliferative lupus nephritis (LN) and its role in predicting treatment response. METHODS Fifty-four biopsy proven proliferative LN patients scheduled to receive high-dose (HD) or low-dose (LD) CYC were recruited and followed up for six months. At six months, patients were classified as clinical responders (CR) or non-responders (NR) to treatment, using the EULAR/EDTA criteria. An IFN-gene based score (IGS) was developed from the mean log-transformed gene expression of MX1, OAS1, IFIT1, OASL, IFIT4, LY6E, IRF7 at baseline, three and six months. Longitudinal changes of IGS within and between groups were assessed and ΔIGS, which is the difference in IGS between baseline and three months was calculated. Independent predictors of non-response were identified and an ROC analysis was performed to evaluate their utility to predict NR. RESULTS There was a dynamic change in IGS within the HD, LD, CR, and NR groups. Compared to baseline, there was a significant decrease in IGS at three months in HD and LD groups (HD group: 2.01 to 1.14, p = .001; LD group = 2.01 to 0.81, p < .001), followed by a significant increase from three to six months in LD group (LD: 0.81 to 1.51, p = .03; HD: 1.14 to 1.54, p = .300). A decrease in IGS from baseline to three months was seen in both CR (2.13 to 0.79, p < .001) and NR groups (1.83 to 1.27, p = .046), and a significant increase from three to six months was observed only in the CR group (CR: 0.79 to 1.57, p = .006; NR: 1.27 to 1.46, p = 1). ΔIGS (baseline to three months) was higher in CR compared to NR group (-1.339 vs -0.563, p = .017). ROC analysis showed that the model comprising of 0.81 fold decrease in IGS from baseline to three months, endocapillary hypercellularity and interstitial inflammation on renal histopathology predicted non-response with a sensitivity of 83.3% and specificity of 71.4%. CONCLUSION In proliferative LN, treated with HD or LD-CYC, combined model comprising of decrease in IGS score by 0.81 fold from baseline to three months, along with important histopathological features such as endocapillary hypercellularity and interstitial inflammation had better predictive capability for non-response.
Collapse
Affiliation(s)
- Sree Nethra Bulusu
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Christina Mary Mariaselvam
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sanket Shah
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Vallayyachari Kommoju
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Chengappa Kavadichanda
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | | | - Molly Thabah
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Vir Singh Negi
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
4
|
Pan L, Liu J, Liu C, Guo L, Yang S. Intermittent pulses of methylprednisolone with low-dose prednisone attenuate lupus symptoms in B6.MRL-Fas lpr/J mice with fewer glucocorticoid side effects. Biomed Pharmacother 2024; 177:117138. [PMID: 39018878 DOI: 10.1016/j.biopha.2024.117138] [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: 05/10/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory and immunosuppressant medications and remain the cornerstone of systemic lupus erythematosus (SLE) therapy. However, ongoing exposure to GCs has the potential to elicit multiple adverse effects. Considering the irreplaceability of GCs in SLE therapy, it is important to explore the optimal regimen of GCs. Here, we compared the long-term efficacy and safety of pulsed and oral GC therapy in a lupus-prone mouse model. Mice were grouped using a randomized block design. We monitored survival rates, proteinuria, serum autoantibodies, and complement 3 (C3) levels up to 28 weeks of age, and assessed renal damage, bone quality, lipid deposition in the liver and marrow, glucose metabolic parameters, and levels of hormones of the hypothalamic-pituitary-adrenal (HPA) axis. Finally, we explored the mechanisms underlying the superior efficacy of the pulse regimen over oral prednisone regimen. We found that both GC regimens alleviated the poor survival rate, proteinuria, and glomerulonephritis, while also reducing serum autoantibodies and increasing the level of C3. The pulsed GC regimen showed less resistance to insulin, less suppression of the HPA axis, less bone loss, and less bone marrow fat deposition than the oral GC regimen. Additionally, GC-induced leucine zipper (GILZ) was significantly overexpressed in the GC pulse group. These results suggest that the GC pulse regimen ameliorated symptoms in lupus-prone mice, with fewer side effects, which may be related to GILZ overexpression. Our findings offer a potentially promising GC treatment option for SLE.
Collapse
Affiliation(s)
- Lu Pan
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China
| | - Jinxiang Liu
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Congcong Liu
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Lishuang Guo
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China
| | - Sirui Yang
- Department of Pediatric Rheumatology, Immunology & Allergy, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; The Child Health Clinical Research Center of Jilin Province, China.
| |
Collapse
|
5
|
Jones SA, Morand EF. Targeting Interferon Signalling in Systemic Lupus Erythematosus: Lessons Learned. Drugs 2024; 84:625-635. [PMID: 38807010 PMCID: PMC11196297 DOI: 10.1007/s40265-024-02043-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
The development of new medicines for systemic lupus erythematosus (SLE) has not addressed unmet clinical need, with only three drugs receiving regulatory approval for SLE in the last 60 years, one of which was specifically licensed for lupus nephritis. In the last 20 years it has become clear that activation of type 1 interferons (IFN) is reproducibly detected in the majority of SLE patients, and the actions of IFN in the immune system and on target tissues is consistent with a pathogenic role in SLE. These findings led to considerable drug discovery activity, first with agents directly targeting IFN family cytokines, with results that were encouraging but underwhelming. In contrast, targeting the type I IFN receptor with the monoclonal antibody anifrolumab, thereby blocking all IFN family members, was effective in a phase II clinical trial. This led to a pair of phase III trials, one of which was negative and the other positive, reflecting the difficulty of obtaining outcomes from trials in this complex disease. Nonetheless, the balance of evidence resulted in approval of anifrolumab in multiple jurisdictions from 2021 onwards. Multiple approaches to targeting the type 1 IFN pathway have subsequently had positive phase II clinical trials, including antibodies targeting cells that produce IFN, and small molecules targeting the receptor kinase TYK2, required for IFN signalling. Despite multiple hurdles, it is clear that IFN targeting in SLE is here to stay. The story of IFN-targeting therapy in SLE has lessons for drug development overall in this disease.
Collapse
Affiliation(s)
- Sarah A Jones
- Centre for Inflammatory Disease, Monash University, Clayton, Australia
| | - Eric F Morand
- Centre for Inflammatory Disease, Monash University, Clayton, Australia.
- Department of Rheumatology, Monash Health, Melbourne, Australia.
- Monash Medical Centre, 246 Clayton Rd, Clayton, VIC, 3168, Australia.
| |
Collapse
|
6
|
Tsoi A, Nikolopoulos D, Parodis I. Advances in the pharmacological management of systemic lupus erythematosus. Expert Opin Pharmacother 2024; 25:705-716. [PMID: 38756102 DOI: 10.1080/14656566.2024.2354457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Despite setbacks in clinical trials for systemic lupus erythematosus (SLE), three drugs have been approved for SLE and lupus nephritis (LN) treatment in the past decade. Several ongoing clinical trials, some viewed optimistically by the scientific community, underscore the evolving landscape. Emerging clinical data have established specific therapeutic targets in routine clinical practice for treating SLE, aiming to improve long-term outcomes. AREAS COVERED Research related to treatment of SLE and LN is discussed, focusing on randomized clinical trials during the last 5 years and recommendations for the management of SLE published by the European Alliance of Associations for Rheumatology (EULAR), American College of Rheumatology (ACR), Asia Pacific League of Associations for Rheumatology (APLAR), and Pan-American League of Associations of Rheumatology (PANLAR). EXPERT OPINION The landscape of SLE and LN treatments is evolving, as new drugs and combination treatment approaches redefine the traditional concepts of induction and maintenance treatment phases. As the therapeutic armamentarium in SLE continues to expand, the research focus is shifting from the imperative for new therapies to advancing our understanding of optimal treatment selection for individual patients, steering toward precision medicine strategies.
Collapse
Affiliation(s)
- Alexander Tsoi
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Dionysis Nikolopoulos
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
7
|
Shirahama Y, Hashimoto A, Ono N, Takeyama Y, Maruyama A, Inoue T, Tada Y, Niiro H. Relationships between Type 1 interferon signatures and clinical features of the new-onset lupus patients in Japan. Mod Rheumatol 2024; 34:346-351. [PMID: 36695430 DOI: 10.1093/mr/road015] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The objective of the study is to investigate the relationships between Type 1 interferon (T1-IFN) signatures and clinical characteristics of lupus patients. METHODS We examined 49 new-onset lupus patients who were diagnosed between 1999 and 2017. The patients treated with >10 mg of prednisolone or hydroxychloroquine were excluded from this study. Serum T1-IFN signatures were revealed by a functional reporter assay and standardized by recombinant IFN-α. Patient backgrounds, clinical findings, and treatments were retrospectively extracted from their electrical medical records. Clinical data were also available, including SLE Disease Activity Index of SLE patients on admission. RESULTS T1-IFN signatures of lupus patients closely correlated with lupus disease activities, such as SLE Disease Activity Index-2K, white blood cell, C3 levels, and the titre of double-strand DNA antibody. We found fever and acute lupus dermatitis closely associated with T1-IFN signature. CONCLUSIONS In lupus patients, fever and acute lupus dermatitis are good indicators of a strong T1-IFN signature.
Collapse
Affiliation(s)
- Yuri Shirahama
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Aki Hashimoto
- Department of Dermatology, Saga University Hospital, Saga, Japan
| | - Nobuyuki Ono
- Department of Rheumatology, Saga University Hospital, Saga, Japan
- Department of Clinical Immunology and Rheumatology/Infectious Disease, Kyushu University Hospital, Fukuoka, Japan
| | - Yukiko Takeyama
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Akihito Maruyama
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Takuya Inoue
- Department of Dermatology, Saga University Hospital, Saga, Japan
| | - Yoshifumi Tada
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Hiroaki Niiro
- Department of Clinical Immunology and Rheumatology/Infectious Disease, Kyushu University Hospital, Fukuoka, Japan
- Department of Medical Education, Graduate School of Medical Sciences, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Deshmukh A, Pereira A, Geraci N, Tzvetkov E, Przetak M, Catalina MD, Morand EF, Bender AT, Vaidyanathan B. Preclinical Evidence for the Glucocorticoid-Sparing Potential of a Dual Toll-Like Receptor 7/8 Inhibitor in Autoimmune Diseases. J Pharmacol Exp Ther 2024; 388:751-764. [PMID: 37673681 DOI: 10.1124/jpet.123.001744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023] Open
Abstract
Toll-like receptor 7 (TLR7) and TLR8 are single-stranded RNA-sensing endosomal pattern recognition receptors that evolved to defend against viral infections. However, aberrant TLR7/8 activation by endogenous ligands has been implicated in the pathogenesis of autoimmune diseases including systemic lupus erythematosus. TLR activation and type I interferon (IFN) were shown recently to impart resistance to glucocorticoids (GC), which are part of the standard of care for multiple autoimmune diseases. While GCs are effective, a plethora of undesirable effects limit their use. New treatment approaches that allow for the use of lower and safer doses of GCs would be highly beneficial. Herein, we report that a dual TLR7/8 inhibitor (TLR7/8i) increases the effectiveness of GCs in inflammatory settings. Human peripheral blood mononuclear cell studies revealed increased GC sensitivity in the presence of TLR7/8i for reducing inflammatory cytokine production, a synergistic effect that was most pronounced in myeloid cells, particularly monocytes. Gene expression analysis by NanoString and single-cell RNA sequencing revealed that myeloid cells were substantially impacted by combining low-dose TLR7/8i and GC, as evidenced by the effects on nuclear factor-kappa B-regulated cytokines and GC-response genes, although IFNs were affected to a smaller degree. Low dose of TLR7/8i plus GC was more efficacious then either agent alone in the MRL/lpr mouse model of lupus, with improved proteinuria and survival. Overall, our findings indicate a GC-sparing potential for TLR7/8i compounds, suggesting TLR7/8i may offer a new strategy for the treatment of autoimmune diseases. SIGNIFICANCE STATEMENT: Some features of autoimmune diseases may be resistant to glucocorticoids, mediated at least in part by toll-like receptor (TLR) activation, necessitating higher doses that are associated with considerable toxicities. We demonstrate that TLR7/8 inhibition and glucocorticoids work synergistically to reduce inflammation in a cell-type specific manner and suppress disease in a mouse model of lupus. TLR7/8 inhibition is a promising strategy for the treatment of autoimmune diseases and has glucocorticoid-sparing potential.
Collapse
Affiliation(s)
- Ankita Deshmukh
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Albertina Pereira
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Nicholas Geraci
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Evgeni Tzvetkov
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Melinda Przetak
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Michelle D Catalina
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Eric F Morand
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Andrew T Bender
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| | - Bharat Vaidyanathan
- Research Unit - Neuroscience and Immunology, EMD Serono, Billerica, Massachusetts (A.D., A.P., N.G., E.T., M.P., M.D.C., A.T. B., B.V.) and School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia (E.F.M.)
| |
Collapse
|
9
|
Xie Z, Chen C, Ma’ayan A. Dex-Benchmark: datasets and code to evaluate algorithms for transcriptomics data analysis. PeerJ 2023; 11:e16351. [PMID: 37953774 PMCID: PMC10638921 DOI: 10.7717/peerj.16351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023] Open
Abstract
Many tools and algorithms are available for analyzing transcriptomics data. These include algorithms for performing sequence alignment, data normalization and imputation, clustering, identifying differentially expressed genes, and performing gene set enrichment analysis. To make the best choice about which tools to use, objective benchmarks can be developed to compare the quality of different algorithms to extract biological knowledge maximally and accurately from these data. The Dexamethasone Benchmark (Dex-Benchmark) resource aims to fill this need by providing the community with datasets and code templates for benchmarking different gene expression analysis tools and algorithms. The resource provides access to a collection of curated RNA-seq, L1000, and ChIP-seq data from dexamethasone treatment as well as genetic perturbations of its known targets. In addition, the website provides Jupyter Notebooks that use these pre-processed curated datasets to demonstrate how to benchmark the different steps in gene expression analysis. By comparing two independent data sources and data types with some expected concordance, we can assess which tools and algorithms best recover such associations. To demonstrate the usefulness of the resource for discovering novel drug targets, we applied it to optimize data processing strategies for the chemical perturbations and CRISPR single gene knockouts from the L1000 transcriptomics data from the Library of Integrated Network Cellular Signatures (LINCS) program, with a focus on understudied proteins from the Illuminating the Druggable Genome (IDG) program. Overall, the Dex-Benchmark resource can be utilized to assess the quality of transcriptomics and other related bioinformatics data analysis workflows. The resource is available from: https://maayanlab.github.io/dex-benchmark.
Collapse
Affiliation(s)
- Zhuorui Xie
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clara Chen
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Ma’ayan
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Abstract
Systemic lupus erythematosus (SLE) is a severe multisystem autoimmune disease that can cause injury in almost every body system. While considered a classic example of autoimmunity, it is still relatively poorly understood. Treatment with immunosuppressive agents is challenging, as many agents are relatively non-specific, and the underlying disease is characterized by unpredictable flares and remissions. This State of The Art Review provides a comprehensive current summary of systemic lupus erythematosus based on recent literature. In basic and translational science, this summary includes the current state of genetics, epigenetics, differences by ancestry, and updates about the molecular and immunological pathogenesis of systemic lupus erythematosus. In clinical science, the summary includes updates in diagnosis and classification, clinical features and subphenotypes, and current guidelines and strategies for treatment. The paper also provides a comprehensive review of the large number of recent clinical trials in systemic lupus erythematosus. Current knowns and unknowns are presented, and potential directions for the future are suggested. Improved knowledge of immunological pathogenesis and the molecular differences that exist between patients should help to personalize treatment, minimize side effects, and achieve better outcomes in this difficult disease.
Collapse
Affiliation(s)
- Eric F Morand
- School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Department of Rheumatology, Monash Health, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
11
|
Tanaka Y, Kusuda M, Yamaguchi Y. Interferons and systemic lupus erythematosus: Pathogenesis, clinical features, and treatments in interferon-driven disease. Mod Rheumatol 2023; 33:857-867. [PMID: 36440704 DOI: 10.1093/mr/roac140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023]
Abstract
Type I interferons (IFNs) have recently received a lot of attention with the elucidation of the pathogenesis of systemic lupus erythematosus (SLE). Type I IFNs are associated with many SLE symptoms and play a role in the pathogenesis of autoimmune diseases that may occur concurrently with SLE, such as Sjögren's syndrome, antiphospholipid syndrome, myositis, scleroderma, and interferonopathy. Type I IFNs could be the link between these diseases. However, direct measurement of type I IFN levels and the IFN gene signature is currently unavailable in clinical practice. This review discusses type I IFN signalling in SLE, investigates the role of type I IFN in the clinical manifestations and symptoms associated with SLE and other IFN-related diseases, and discusses the clinical tests that can be used to diagnose SLE and measure disease activity. In addition, the role of type I IFN-blocking therapies as potential treatments for SLE is discussed.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | |
Collapse
|
12
|
Fasano S, Milone A, Nicoletti GF, Isenberg DA, Ciccia F. Precision medicine in systemic lupus erythematosus. Nat Rev Rheumatol 2023; 19:331-342. [PMID: 37041269 DOI: 10.1038/s41584-023-00948-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/13/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that has diverse clinical manifestations, ranging from restricted cutaneous involvement to life-threatening systemic organ involvement. The heterogeneity of pathomechanisms that lead to SLE contributes to between-patient variation in clinical phenotype and treatment response. Ongoing efforts to dissect cellular and molecular heterogeneity in SLE could facilitate the future development of stratified treatment recommendations and precision medicine, which is a considerable challenge for SLE. In particular, some genes involved in the clinical heterogeneity of SLE and some phenotype-related loci (STAT4, IRF5, PDGF genes, HAS2, ITGAM and SLC5A11) have an association with clinical features of the disease. An important part is also played by epigenetic varation (in DNA methylation, histone modifications and microRNAs) that influences gene expression and affects cell function without modifying the genome sequence. Immune profiling can help to identify an individual's specific response to a therapy and can potentially predict outcomes, using techniques such as flow cytometry, mass cytometry, transcriptomics, microarray analysis and single-cell RNA sequencing. Furthermore, the identification of novel serum and urinary biomarkers would enable the stratification of patients according to predictions of long-term outcomes and assessments of potential response to therapy.
Collapse
Affiliation(s)
- Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Alessandra Milone
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - David A Isenberg
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
13
|
Bruce IN, van Vollenhoven RF, Morand EF, Furie RA, Manzi S, White WB, Abreu G, Tummala R. Sustained glucocorticoid tapering in the phase 3 trials of anifrolumab: a post hoc analysis of the TULIP-1 and TULIP-2 trials. Rheumatology (Oxford) 2023; 62:1526-1534. [PMID: 36018235 PMCID: PMC10070065 DOI: 10.1093/rheumatology/keac491] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES Glucocorticoid sparing is a key priority for SLE management. We evaluated the effects of sustained glucocorticoid tapering in patients with SLE. MATERIAL AND METHODS This was a post hoc analysis of the randomized, placebo-controlled, 52-week phase 3 Treatment of Uncontrolled Lupus via the Interferon Pathway (TULIP)-1 and TULIP-2 trials of anifrolumab (300 mg i.v. once every 4 weeks for 48 weeks) plus standard therapy in patients with moderate to severe SLE. In a cohort of patients receiving glucocorticoids (prednisone or equivalent) 10 mg or more per day at baseline, we assessed changes in glucocorticoid dosage, patient-reported outcomes (PROs) and safety. Outcome measures were compared between sustained glucocorticoid taper responders (7.5 mg or less per day by week 40 sustained through week 52) and non-responders, regardless of treatment group, and between patients receiving anifrolumab or placebo. RESULTS Among the 726 patients in the TULIP trials, 375 patients received glucocorticoids 10 mg or more per day at baseline, and of these, 155 (41%) patients were sustained glucocorticoid taper responders. Compared with non-responders (n = 220), sustained glucocorticoid taper responders reduced their mean cumulative glucocorticoid dose by 32%, improved PRO scores, reduced blood pressure and experienced fewer serious adverse events. Sustained glucocorticoid tapering was achieved by 51% (96/190) of patients receiving anifrolumab vs 32% (59/185) receiving placebo. Compared with placebo, more anifrolumab-treated patients achieved both sustained glucocorticoid taper and reduced overall disease activity [38% (72/190) vs 23% (43/185)]. CONCLUSIONS Sustained glucocorticoid tapering is associated with clinical benefits. Anifrolumab treatment has potential to reduce disease activity and glucocorticoid exposure, a key goal of SLE management. STUDY REGISTRATION ClinicalTrials.gov identifier: NCT02446912 and NCT02446899.
Collapse
Affiliation(s)
- Ian N Bruce
- Centre for Epidemiology Versus Arthritis, The University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Ronald F van Vollenhoven
- Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric F Morand
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Richard A Furie
- Division of Rheumatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Susan Manzi
- Lupus Center of Excellence, Autoimmunity Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - William B White
- Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Gabriel Abreu
- BioPharmaceuticals R&D, AstraZeneca R&D, Gothenburg, Sweden
| | - Raj Tummala
- BioPharmaceuticals R&D, AstraZeneca US, Gaithersburg, MD, USA
| |
Collapse
|
14
|
Dankers W, Northcott M, Bennett T, D’Cruz A, Sherlock R, Gearing LJ, Hertzog P, Russ B, Miceli I, Scheer S, Fujishiro M, Hayakawa K, Ikeda K, Morand EF, Jones SA. Type 1 interferon suppresses expression and glucocorticoid induction of glucocorticoid-induced leucine zipper (GILZ). Front Immunol 2022; 13:1034880. [PMID: 36505447 PMCID: PMC9727222 DOI: 10.3389/fimmu.2022.1034880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022] Open
Abstract
SLE is a systemic multi-organ autoimmune condition associated with reduced life expectancy and quality of life. Glucocorticoids (GC) are heavily relied on for SLE treatment but are associated with detrimental metabolic effects. Type 1 interferons (IFN) are central to SLE pathogenesis and may confer GC insensitivity. Glucocorticoid-induced leucine zipper (GILZ) mediates many effects of GC relevant to SLE pathogenesis, but the effect of IFN on GC regulation of GILZ is unknown. We performed in vitro experiments using human PBMC to examine the effect of IFN on GILZ expression. JAK inhibitors tofacitinib and tosylate salt were used in vivo and in vitro respectively to investigate JAK-STAT pathway dependence of our observations. ChiP was performed to examine glucocorticoid receptor (GR) binding at the GILZ locus. Several public data sets were mined for correlating clinical data. High IFN was associated with suppressed GILZ and reduced GILZ relevant to GC exposure in a large SLE population. IFN directly reduced GILZ expression and suppressed the induction of GILZ by GC in vitro in human leukocytes. IFN actions on GILZ expression were dependent on the JAK1/Tyk2 pathway, as evidenced by loss of the inhibitory effect of IFN on GILZ in the presence of JAK inhibitors. Activation of this pathway led to reduced GR binding in key regulatory regions of the GILZ locus. IFN directly suppresses GILZ expression and GILZ upregulation by GC, indicating a potential mechanism for IFN-induced GC resistance. This work has important implications for the ongoing development of targeted GC-sparing therapeutics in SLE.
Collapse
Affiliation(s)
- Wendy Dankers
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Melissa Northcott
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Taylah Bennett
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Akshay D’Cruz
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Rochelle Sherlock
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Linden J. Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Paul Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Brendan Russ
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Iolanda Miceli
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Sebastian Scheer
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Maki Fujishiro
- Institutes for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Kunihiro Hayakawa
- Institutes for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Keigo Ikeda
- Institutes for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
- Department of Internal Medicine and Rheumatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Eric F. Morand
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| | - Sarah A. Jones
- Centre for Inflammatory Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Wahadat MJ, Schonenberg-Meinema D, van Helden-Meeuwsen CG, van Tilburg SJ, Groot N, Schatorjé EJH, Hoppenreijs EPAH, Hissink Muller PCE, Brinkman DMC, Dvorak D, Verkaaik M, van den Berg JM, Bouchalova K, Kamphuis S, Versnel MA. Gene signature fingerprints stratify SLE patients in groups with similar biological disease profiles: a multicentre longitudinal study. Rheumatology (Oxford) 2022; 61:4344-4354. [PMID: 35143620 PMCID: PMC9629374 DOI: 10.1093/rheumatology/keac083] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/31/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Clinical phenotyping and predicting treatment responses in SLE patients is challenging. Extensive blood transcriptional profiling has identified various gene modules that are promising for stratification of SLE patients. We aimed to translate existing transcriptomic data into simpler gene signatures suitable for daily clinical practice. METHODS Real-time PCR of multiple genes from the IFN M1.2, IFN M5.12, neutrophil (NPh) and plasma cell (PLC) modules, followed by a principle component analysis, was used to identify indicator genes per gene signature. Gene signatures were measured in longitudinal samples from two childhood-onset SLE cohorts (n = 101 and n = 34, respectively), and associations with clinical features were assessed. Disease activity was measured using Safety of Estrogen in Lupus National Assessment (SELENA)-SLEDAI. Cluster analysis subdivided patients into three mutually exclusive fingerprint-groups termed (1) all-signatures-low, (2) only IFN high (M1.2 and/or M5.12) and (3) high NPh and/or PLC. RESULTS All gene signatures were significantly associated with disease activity in cross-sectionally collected samples. The PLC-signature showed the highest association with disease activity. Interestingly, in longitudinally collected samples, the PLC-signature was associated with disease activity and showed a decrease over time. When patients were divided into fingerprints, the highest disease activity was observed in the high NPh and/or PLC group. The lowest disease activity was observed in the all-signatures-low group. The same distribution was reproduced in samples from an independent SLE cohort. CONCLUSIONS The identified gene signatures were associated with disease activity and were indicated to be suitable tools for stratifying SLE patients into groups with similar activated immune pathways that may guide future treatment choices.
Collapse
Affiliation(s)
- M Javad Wahadat
- Department of Immunology, Erasmus MC
- Department of Paediatric Rheumatology, Erasmus MC—Sophia Children’s hospital, University Medical Center Rotterdam, Rotterdam
| | - Dieneke Schonenberg-Meinema
- Department of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children's Hospital, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam
| | | | | | - Noortje Groot
- Department of Paediatric Rheumatology, Erasmus MC—Sophia Children’s hospital, University Medical Center Rotterdam, Rotterdam
| | - Ellen J H Schatorjé
- Department of Paediatric Rheumatology, Amalia Children’s Hospital, Radboudumc
- Department of Paediatric Rheumatology, St. Maartenskliniek, Nijmegen
| | - Esther P A H Hoppenreijs
- Department of Paediatric Rheumatology, Amalia Children’s Hospital, Radboudumc
- Department of Paediatric Rheumatology, St. Maartenskliniek, Nijmegen
| | - Petra C E Hissink Muller
- Department of Pediatrics, Division of Pediatric Rheumatology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Danielle M C Brinkman
- Department of Pediatrics, Division of Pediatric Rheumatology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Denis Dvorak
- Paediatric Rheumatology, Department of Paediatrics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Marleen Verkaaik
- Department of Paediatric Rheumatology, Erasmus MC—Sophia Children’s hospital, University Medical Center Rotterdam, Rotterdam
| | - J Merlijn van den Berg
- Department of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children's Hospital, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam
| | - Kateřina Bouchalova
- Paediatric Rheumatology, Department of Paediatrics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Sylvia Kamphuis
- Department of Paediatric Rheumatology, Erasmus MC—Sophia Children’s hospital, University Medical Center Rotterdam, Rotterdam
| | | |
Collapse
|
16
|
Nataraja C, Flynn J, Dankers W, Northcott M, Zhu W, Sherlock R, Bennett TJ, Russ BE, Miceli I, Pervin M, D'Cruz A, Harris J, Morand EF, Jones SA. GILZ regulates type I interferon release and sequesters STAT1. J Autoimmun 2022; 131:102858. [PMID: 35810690 DOI: 10.1016/j.jaut.2022.102858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022]
Abstract
Glucocorticoids remain a mainstay of modern medicine due to their ability to broadly suppress immune activation. However, they cause severe adverse effects that warrant urgent development of a safer alternative. The glucocorticoid-induced leucine zipper (GILZ) gene, TSC22D3, is one of the most highly upregulated genes in response to glucocorticoid treatment, and reduced GILZ mRNA and protein levels are associated with increased severity of inflammation in systemic lupus erythematosus (SLE), Ulcerative Colitis, Psoriasis, and other autoimmune/autoinflammatory diseases. Here, we demonstrate that low GILZ permits expression of a type I interferon (IFN) signature, which is exacerbated in response to TLR7 and TLR9 stimulation. Conversely, overexpression of GILZ prevents IFN-stimulated gene (ISG) up-regulation in response to IFNα. Moreover, GILZ directly binds STAT1 and prevents its nuclear translocation, thereby negatively regulating IFN-induced gene expression and the auto-amplification loop of the IFN response. Thus, GILZ powerfully regulates both the expression and action of type I IFN, suggesting restoration of GILZ as an attractive therapeutic strategy for reducing reliance on glucocorticoids.
Collapse
Affiliation(s)
- Champa Nataraja
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Jacqueline Flynn
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Wendy Dankers
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Melissa Northcott
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Wendy Zhu
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Rochelle Sherlock
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Taylah J Bennett
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Brendan E Russ
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Iolanda Miceli
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Mehnaz Pervin
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Akshay D'Cruz
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - James Harris
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Eric F Morand
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia
| | - Sarah A Jones
- Rheumatology Research Group, Monash University Centre for Inflammatory Disease, School of Clinical Sciences at Monash Health, 246 Clayton Rd, Clayton, 3168, Melbourne, Australia.
| |
Collapse
|
17
|
Chalhoub NE, Wenderfer SE, Levy DM, Rouster-Stevens K, Aggarwal A, Savani SI, Ruth NM, Arkachaisri T, Qiu T, Merritt A, Onel K, Goilav B, Khubchandani RP, Deng J, Fonseca AR, Ardoin SP, Ciurtin C, Kasapcopur O, Jelusic M, Huber AM, Ozen S, Klein-Gitelman MS, Appenzeller S, Cavalcanti A, Fotis L, Lim SC, Silva RM, Miramontes JR, Rosenwasser NL, Saad-Magalhaes C, Schonenberg-Meinema D, Scott C, Silva CA, Enciso S, Terreri MT, Torres-Jimenez AR, Trachana M, Al-Mayouf SM, Devarajan P, Huang B, Brunner HI. International Consensus for the Dosing of Corticosteroids in Childhood-Onset Systemic Lupus Erythematosus With Proliferative Lupus Nephritis. Arthritis Rheumatol 2022; 74:263-273. [PMID: 34279063 PMCID: PMC8766607 DOI: 10.1002/art.41930] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/15/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To develop a standardized steroid dosing regimen (SSR) for physicians treating childhood-onset systemic lupus erythematosus (SLE) complicated by lupus nephritis (LN), using consensus formation methodology. METHODS Parameters influencing corticosteroid (CS) dosing were identified (step 1). Data from children with proliferative LN were used to generate patient profiles (step 2). Physicians rated changes in renal and extrarenal childhood-onset SLE activity between 2 consecutive visits and proposed CS dosing (step 3). The SSR was developed using patient profile ratings (step 4), with refinements achieved in a physician focus group (step 5). A second type of patient profile describing the course of childhood-onset SLE for ≥4 months since kidney biopsy was rated to validate the SSR-recommended oral and intravenous (IV) CS dosages (step 6). Patient profile adjudication was based on majority ratings for both renal and extrarenal disease courses, and consensus level was set at 80%. RESULTS Degree of proteinuria, estimated glomerular filtration rate, changes in renal and extrarenal disease activity, and time since kidney biopsy influenced CS dosing (steps 1 and 2). Considering these parameters in 5,056 patient profile ratings from 103 raters, and renal and extrarenal course definitions, CS dosing rules of the SSR were developed (steps 3-5). Validation of the SSR for up to 6 months post-kidney biopsy was achieved with 1,838 patient profile ratings from 60 raters who achieved consensus for oral and IV CS dosage in accordance with the SSR (step 6). CONCLUSION The SSR represents an international consensus on CS dosing for use in patients with childhood-onset SLE and proliferative LN. The SSR is anticipated to be used for clinical care and to standardize CS dosage during clinical trials.
Collapse
Affiliation(s)
| | | | - Deborah M Levy
- The Hospital for Sick Children and The University of Toronto, Toronto, Ontario, Canada
| | | | - Amita Aggarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | | | - Tingting Qiu
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Angela Merritt
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Karen Onel
- Hospital for Special Surgery, New York, New York
| | | | | | - Jianghong Deng
- Capital Medical University and National Center for Children's Health, Beijing, China
| | | | | | | | - Ozgur Kasapcopur
- Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Marija Jelusic
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Adam M Huber
- IWK Health Centre and Dalhousie University, Halifax, Nova Scotia, Canada
| | - Seza Ozen
- Hacettepe University, Ankara, Turkey
| | - Marisa S Klein-Gitelman
- Northwestern University Feinberg School of Medicine and Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | | | - André Cavalcanti
- Hospital das Clínicas da Universidade Federal de Pernambuco, Recife, Brazil
| | - Lampros Fotis
- National and Kapodistian University of Athens, Athens, Greece
| | | | - Rodrigo M Silva
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Christiaan Scott
- Red Cross War Memorial Children's Hospital and University of Cape Town, Cape Town, South Africa
| | | | - Sandra Enciso
- Hospital de la Beneficencia Española, Mexico City, Mexico
| | | | | | - Maria Trachana
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sulaiman M Al-Mayouf
- King Faisal Specialist Hospital and Research Center and Alfaisal University, Riyadh, Saudi Arabia
| | - Prasad Devarajan
- University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bin Huang
- University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hermine I Brunner
- University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
18
|
Northcott M, Jones S, Koelmeyer R, Bonin J, Vincent F, Kandane-Rathnayake R, Hoi A, Morand E. Type 1 interferon status in systemic lupus erythematosus: a longitudinal analysis. Lupus Sci Med 2022; 9:e000625. [PMID: 35197305 PMCID: PMC8867321 DOI: 10.1136/lupus-2021-000625] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/04/2022] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Type 1 interferon (IFN) is key to the pathogenesis of SLE, evidenced by the expression of IFN-stimulated genes (ISGs) in most patients, but the clinical utility of serial ISG assessment remains unknown. With the emergence of IFN-blocking drugs, we aimed to examine IFN status in relation to clinical findings longitudinally to provide insights into the value of testing ISG levels over time. METHODS Clinical data and whole blood were collected prospectively on adult patients with SLE from a single tertiary lupus centre. IFN status was measured using a panel of ISGs. FINDINGS 729 samples were analysed from 205 patients. At baseline, 62.9% of patients were IFN high, 30.2% IFN low and 6.8% borderline. 142 patients had multiple samples collected, and 87.3% of these demonstrated stable ISG status over time. In longitudinal follow-up, IFN high patients had higher activity in multiple organ domains and spent less time in Lupus Low Disease Activity State, but IFN score did not correlate with SLE Disease Activity Index in individual patients. In the small subset of patients who had large fluctuations in ISG across the observation period, most had high-dose glucocorticoids that correlated with ISG suppression. However, low-moderate-dose glucocorticoids did not suppress ISG expression. CONCLUSION Although IFN high status is associated with indicators of more severe SLE, in the majority of patients, ISGs are stable across time and do not correlate with disease activity. Changes in ISG expression may be seen with high-dose, but not routine dose, glucocorticoid exposure. These findings suggest baseline but not serial ISG measurement may be of value in the management of SLE.
Collapse
Affiliation(s)
- Melissa Northcott
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Sarah Jones
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rachel Koelmeyer
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Julie Bonin
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Fabien Vincent
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Rangi Kandane-Rathnayake
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Alberta Hoi
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Eric Morand
- Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|