1
|
Wu C, Jiang S, Chen Z, Li T, Gu X, Dai M, Du F, Ye Y, Tang L, Wang M, Wang X, Li T, Ye S, Bao C, Zhang X, Fu Q. Single-cell transcriptomics reveal potent extrafollicular B cell response linked with granzyme K + CD8 T cell activation in lupus kidney. Ann Rheum Dis 2024:ard-2024-225876. [PMID: 39419536 DOI: 10.1136/ard-2024-225876] [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: 03/27/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES B and T cells constitute the majority of infiltrating lymphocytes in the kidney and represent the local perpetrators in lupus nephritis (LN), but the underlying pathogenic mechanisms are not well elucidated. The aim of this study is to explore the kidney-specific adaptive immune landscape in patients with active LN at the single-cell level. METHODS We performed single-cell RNA/B cell receptor (BCR)/T cell receptor (TCR) sequencing analysis on sorting-purified B and T cells from the kidney and paired peripheral blood of patients with active LN, and the periphery of matched controls. Flow cytometry, Assay for Transposase Accessible-sequencing, multiplexed immunohistochemistry and functional studies were performed to validate the transcriptomic results. RESULTS High infiltrations of intrarenal atypical B cells (ABCs) and antibody-secreting cells (ASCs) were identified in the B cell compartment. The single-cell BCR repertoire analysis revealed strong clonal expansion of intrarenal ASCs dominated by IGHG1 and IGHG3 isotypes, accompanied by lower frequencies of heavy-chain and light-chain somatic mutations, compared with the peripheral ASCs. Notably, a unique expansion of IGHG4-59 and clonal overlap between ABCs and ASCs was found in kidney-specific clonotypes. In the T cell compartment, we identified granzyme K (GZMK)+ CD8 T cells as the dominant kidney-associated T cells which shared inflammation- and stress-related gene pathways with ABCs. Intrarenal GZMK+ CD8 T cells highly expressed IFNG and displayed strong communication with ABCs via the type II interferon (IFN) pathway. Intrarenal GZMK+ CD8 T cells and ABCs were largely co-localised within the tertiary lymphoid structure, and GZMK+ CD8 T cells potentially contributed to the differentiation of ABCs via IFN-γ and interleukin-21. CONCLUSIONS Our study revealed a potent extrafollicular B cell response linked with overactivation of GZMK+ CD8 T cells in the kidney of patients with LN, which may lead to innovative treatments for LN.
Collapse
Affiliation(s)
- Chunmei Wu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Shan Jiang
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zechuan Chen
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Teng Li
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xixi Gu
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Dai
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Du
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | - Xiaodong Wang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Li
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunde Bao
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoming Zhang
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiong Fu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Immune Therapy Institute, Shanghai, China
| |
Collapse
|
2
|
Bhoj PS, Nocito C, Togre NS, Winfield M, Lubinsky C, Khan S, Mogadala N, Seliga A, Unterwald EM, Persidsky Y, Sriram U. Tissue Kallikrein-1 Suppresses Type I Interferon Responses and Reduces Depressive-Like Behavior in the MRL/lpr Lupus-Prone Mouse Model. Int J Mol Sci 2024; 25:10080. [PMID: 39337564 PMCID: PMC11432477 DOI: 10.3390/ijms251810080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Excessive production and response to Type I interferons (IFNs) is a hallmark of systemic lupus erythematosus (SLE). Neuropsychiatric lupus (NPSLE) is a common manifestation of human SLE, with major depression as the most common presentation. Clinical studies have demonstrated that IFNα can cause depressive symptoms. We have shown that the kallikrein-kinin system (KKS) [comprised of kallikreins (Klks) and bradykinins] and angiotensin-converting enzyme inhibitors suppressed Type I IFN responses in dendritic cells from lupus-prone mice and human peripheral blood mononuclear cells. Tissue Klk genes are decreased in patients with lupus, and giving exogenous Klk1 ameliorated kidney pathology in mice. We retro-orbitally administered mouse klk1 gene-carrying adenovirus in the Murphy Roths Large lymphoproliferative (MRL/lpr) lupus-prone mice at early disease onset and analyzed immune responses and depressive-like behavior. Klk1 improved depressive-like behavior, suppressed interferon-responsive genes and neuroinflammation, and reduced plasma IFNα levels and proinflammatory cytokines. Klk1 also reduced IFNAR1 and JAK1 protein expression, important upstream molecules in Type I IFN signaling. Klk1 reduced bradykinin B1 receptor expression, which is known to induce proinflammatory response. Together, these findings suggest that Klk1 may be a potential therapeutic candidate to control IFNα production/responses and other inflammatory responses in SLE and NPSLE.
Collapse
Affiliation(s)
- Priyanka S. Bhoj
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Cassandra Nocito
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Namdev S. Togre
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Cody Lubinsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Sabeeya Khan
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Nikhita Mogadala
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Alecia Seliga
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Ellen M. Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (P.S.B.); (C.N.); (N.S.T.); (M.W.); (C.L.); (S.K.); (N.M.); (A.S.); (Y.P.)
| |
Collapse
|
3
|
Zhou P, Huang M, Hang Y, Liu S, Yao G, Tang X, Xia N, Sun L. Artesunate alleviates Sjögren's Syndrome by inhibiting the interferon-α signaling in plasmacytoid dendritic cells via TLR-MyD88-IRF7. Biomed Pharmacother 2024; 177:116885. [PMID: 38878633 DOI: 10.1016/j.biopha.2024.116885] [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: 02/22/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/28/2024] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disease in which the salivary glands (SGs) and the lacrimal glands (LGs) are affected by lymphocytic infiltration and inflammation. It has been reported that interferon-α (IFN-α) released by plasmacytoid dendritic cells (pDCs) contribute to the pathology of SS, and ART has been shown to effectively ameliorates SS. Despite the current research endeavors, the mechanism of how ART works in the treatment of SS remains to be fully elucidated. Whether ART can treat SS by inhibiting IFN-α remains unclear. This hypothesis was tested both in vivo and in vitro settings during the study. The SS model mice, which were treated with ART, showed amelioration in symptoms related to dryness. RNA-seq analysis revealed strong anti-IFN-α signaling response upon ART treatment. Additional in vitro studies provided further confirmation that the application of ART inhibits the MyD88 protein expression and the nuclear translocation of IRF7. This suggests that the intervention of ART in the TLR-MyD88-IRF7 pathway plays a role in the therapeutic approach for SS. In summary, this study highlighted the therapeutic potential of ART in SS and ART inhibited the IFN-α signaling in pDCs via the TLR-MyD88-IRF7 pathway.
Collapse
Affiliation(s)
- Panpan Zhou
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Mengxi Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Yang Hang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Sha Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Nan Xia
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| |
Collapse
|
4
|
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 2024:1-11. [PMID: 38952099 DOI: 10.1080/25785826.2024.2372918] [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: 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
|
5
|
Drougkas K, Skarlis C, Mavragani C. Type I Interferons in Systemic Autoimmune Rheumatic Diseases: Pathogenesis, Clinical Features and Treatment Options. Mediterr J Rheumatol 2024; 35:365-380. [PMID: 39193187 PMCID: PMC11345602 DOI: 10.31138/mjr.270324.tis] [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: 03/27/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 08/29/2024] Open
Abstract
Type I interferon (IFN) pathway dysregulation plays a crucial role in the pathogenesis of several systemic autoimmune rheumatic diseases (SARDs), including systemic lupus erythematosus (SLE), Sjögren's disease (SjD), systemic sclerosis (SSc), dermatomyositis (DM) and rheumatoid arthritis (RA). Genetic and epigenetic alterations have been involved in dysregulated type I IFN responses in systemic autoimmune disorders. Aberrant type I IFN production and secretion have been associated with distinct clinical phenotypes, disease activity, and severity as well as differentiated treatment responses among SARDs. In this review, we provide an overview of the role of type I IFNs in systemic autoimmune diseases including SLE, RA, SjD, SSc, and DM focusing on pathophysiological, clinical, and therapeutical aspects.
Collapse
Affiliation(s)
- Konstantinos Drougkas
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Skarlis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Fan W, Wei B, Chen X, Zhang Y, Xiao P, Li K, Zhang YQ, Huang J, Leng L, Bucala R. Potential role of RhoA GTPase regulation in type interferon signaling in systemic lupus erythematosus. Arthritis Res Ther 2024; 26:31. [PMID: 38243295 PMCID: PMC10799493 DOI: 10.1186/s13075-024-03263-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormal activation of the type I interferon (IFN) pathway, which results in tissue inflammation and organ damage. We explored the role of the RhoA GTPase in the type I IFN activation pathway to provide a potential basis for targeting GTPase signaling for the treatment of SLE. METHODS Total RNA was extracted from peripheral blood mononuclear cells (PBMCs) of SLE patients and healthy controls, and the mRNA expression levels of RhoA and IFN-stimulated genes were measured by SYBR Green quantitative reverse transcriptase-polymerase chain reaction. IFN-a-stimulated response element (ISRE)-luciferase reporter gene assays and Western blotting were conducted to assess the biologic function of RhoA. An enzyme-linked immunoassay (ELISA) measured C-X-C motif chemokine ligand 10 (CXCL10) protein expression. RESULTS Our studies demonstrate that the expression of RhoA in the PBMCs of SLE subjects was significantly higher than in healthy controls and positively correlated with type I IFN scores and type I IFN-stimulated gene (ISGs) expression levels. SiRNA-mediated knockdown of RhoA and the RhoA/ROCK inhibitor Y27632 reduced the activity of the type I IFN-induced ISRE, the signal transducer and activator of transcription 1 (STAT-1) phosphorylation, and the expression of CXCL10 and 2'-5'-oligoadenylate synthetase 1 (OAS1). Finally, we verified that Y27632 could significantly down-regulate the OAS1 and CXCL10 expression levels in the PBMCs of SLE patients. CONCLUSION Our study shows that RhoA positively regulates the activation of the type I IFN response pathway. Reducing the expression level of RhoA inhibits the abnormal activation of the type I IFN system, and the RhoA/ROCK inhibitor Y27632 decreases aberrant type I IFN signaling in SLE PBMCs, suggesting the possibility of targeting the RhoA GTPase for the treatment of SLE.
Collapse
Affiliation(s)
- Wei Fan
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China.
| | - Bo Wei
- Department of Rheumatology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xuyan Chen
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Yi Zhang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Pingping Xiao
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Kaiyan Li
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Yi Qin Zhang
- Department of Nephrology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Jinmei Huang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
7
|
Fan W, Wei B, Chen X, Zhang Y, Xiao P, Li K, Zhang YQ, Huang J, Leng L, Bucala R. The RhoA GTPase regulates Type I Interferon Signaling in Systemic lupus erythematosus. RESEARCH SQUARE 2023:rs.3.rs-3320841. [PMID: 37790522 PMCID: PMC10543431 DOI: 10.21203/rs.3.rs-3320841/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Objective Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormal activation of the type I interferon (IFN) pathway, which results in tissue inflammation and organ damage. We explored the role of the RhoA GTPase in the type I IFN activation pathway to provide a potential basis for targeting GTPase signaling for the treatment of SLE. Methods Total RNA was extracted from peripheral blood mononuclear cells (PBMCs) of SLE patients and healthy controls, and the mRNA expression levels of RhoA and IFN-stimulated genes were measured by SYBR Green quantitative reverse transcriptase-polymerase chain reaction. IFN-stimulated response element (ISRE)-luciferase reporter gene assays and Western blotting were conducted to asssess the biologic function of RhoA. An Enzyme-Linked Immunoassay (ELISA) measured C-X-C motif chemokine ligand 10(CXCL10)protein expression. Results Our studies demonstrated that the expression of RhoA in the PBMCs of SLE subjects was significantly higher than healthy controls and positively correlated with type I IFN scores and type I IFN-stimulated gene (ISGs) expression levels. SiRNA-mediated knockdown of RhoA and the RhoA/ROCK inhibitor Y27632 reduced the activity of the type I IFN-induced ISRE, the signal transducer and activator of transcription 1 (STAT-1) phosphorylation, and the expression of CXCL10 and 2'-5'-oligoadenylate synthetase 1(OAS1). Finally,we verified that Y27632 could significantly down-regulate the OAS1 and CXCL10 expression levels in PBMCs of SLE patients. Conclusion Our study shows that RhoA positively regulates the activation of the type I IFN response pathway. Reducing the expression level of RhoA inhibits the abnormal activation of the type I IFN system, and the RhoA/ROCK inhibitor Y27632 decreases aberrant type I IFN signaling in SLE PBMCs, suggesting the possibility of targeting the RhoA GTPase for the treatment of SLE.
Collapse
Affiliation(s)
- Wei Fan
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Bo Wei
- Zhongshan Hospital of Xiamen University, Medical College of Xiamen University, Xiamen University
| | - Xuyan Chen
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Yi Zhang
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Pingping Xiao
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Kaiyan Li
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Yi Qin Zhang
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Jinmei Huang
- the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College
| | - Lin Leng
- Yale University School of Medicine
| | | |
Collapse
|
8
|
Nasser M, Wadie M, Farid A, Amir AE. Nailfold capillaroscopy in Egyptian systemic lupus erythematosus (SLE) patients: correlation with demographic features and serum levels of IL 17A and IFNs I. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2023; 50:47. [DOI: 10.1186/s43166-023-00215-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2024] Open
Abstract
Abstract
Background
In SLE patients, cytokines are linked to endothelial cell damage. Nailfold capillaroscopy (NFC) is a simple method for evaluating micro-vascular abnormalities in different connective tissue diseases (CTDs). The study aimed to detect the levels of interleukin 17A (IL 17A), type I interferons (IFNs I) in the serum, and NFC changes in Egyptian SLE patients compared to a control group and to correlate NFC findings with patients’ demographic features and serum levels of IL 17A and IFNs I.
Results
Serum levels of IL 17A, IFN α, and IFN β were significantly higher in SLE patients than in control group (P < 0.0001). About thirty nine patients (73.6%) of the 53 SLE patients showed abnormal NFC changes. Egyptian SLE patients had a high prevalence of the NFC non-specific pattern, with 32 (60.4%) patients showing non-specific changes and 7 (13.2%) patients showing scleroderma pattern, including 3 (5.6%) patients with active scleroderma pattern and 4 (7.55%) patients with late scleroderma pattern. Furthermore, Raynaud’s phenomenon (RP) was observed in 8 (15.1%) SLE patients, with 3 (5.6%) having normal NFC pattern and 5 (9.4%) having scleroderma pattern. All controls (n = 20) showed normal hairpin shape capillaries. Except for SLEDAI (P = 0.03) and the presence of RP (P < 0.0001), there were no significant differences in demographic and laboratory parameters between the three NFC patterns (normal, non-specific, and scleroderma); additionally, NFC score correlated significantly with SLEDAI (P = 0.021).
Conclusion
As a result of the high disease activity, Egyptian SLE patients had elevated serum levels of IL 17A and IFNs I. The most common NFC pattern in Egyptian SLE patients was a non-specific pattern. NFC abnormalities in Egyptian SLE patients were correlated with disease activity but not with patients’ ages, disease duration, or serum levels of IL 17A and IFNs I. SLE patients with scleroderma NFC pattern and RP should be closely followed for the possibility of appearance of anti-U1 RNP antibodies and MCTDS.
Collapse
|
9
|
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: 3.0] [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
|
10
|
Korsten P, Tampe B. A Transcriptome Array-Based Approach Links Proteinuria and Distinct Molecular Signatures to Intrarenal Expression of Type I Interferon IFNA5 in Lupus Nephritis. Int J Mol Sci 2023; 24:10636. [PMID: 37445814 DOI: 10.3390/ijms241310636] [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: 05/13/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
In systemic lupus erythematosus (SLE), the relevance of non-hematopoietic sources of type I interferon in human autoimmunity has recently been recognized. Particularly, type I interferon production precedes autoimmunity in early skin lesions related to SLE. However, the relevance of intrarenal type I interferon expression has not been shown in lupus nephritis. From transcriptome array datasets, median-centered log2 mRNA expression levels of IFNα (IFNA1, IFNA2, IFNA4, IFNA5, IFNA6, IFNA7, IFNA8, IFNA10, IFNA13, IFNA14, IFNA16, IFNA17, and IFNA21), IFNω (IFNW1), and IFNβ (IFNB1) in lupus nephritis were extracted specifically from microdissected tubulointerstitial (n = 32) and glomerular compartments (n = 32). We found an association between proteinuria and tubulointerstitial expression of type I interferon IFNA5 (p = 0.0142), while all others were not significantly associated. By contrast, no such correlation was observed between proteinuria and any type I interferon expression in the glomerular compartment in lupus nephritis. Interestingly, there was no difference between female and male patients (p = 0.8237) and no association between type I interferon IFNA5 expression and kidney function or lupus nephritis progression. Finally, we identified distinct molecular signatures involved in transcriptional regulation (GLI protein-regulated transcription, IRF7 activation, and HSF1-dependent transactivation) and receptor signaling (BMP signaling and GPCR ligand binding) in association with tubulointerstitial expression of type I interferon IFNA5 in the kidney. In summary, this transcriptome array-based approach links proteinuria to the tubulointerstitial expression of type I interferon IFNA5 in lupus nephritis. Because type I interferon receptor subunit I antagonism has recently been investigated in active SLE, the current study further emphasizes the role of type I interferons in lupus nephritis and might also be of relevance for mechanistic studies.
Collapse
Affiliation(s)
- Peter Korsten
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
11
|
Mohan C, Zhang T, Putterman C. Pathogenic cellular and molecular mediators in lupus nephritis. Nat Rev Nephrol 2023:10.1038/s41581-023-00722-z. [PMID: 37225921 DOI: 10.1038/s41581-023-00722-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/26/2023]
Abstract
Kidney involvement in patients with systemic lupus erythematosus - lupus nephritis (LN) - is one of the most important and common clinical manifestations of this disease and occurs in 40-60% of patients. Current treatment regimens achieve a complete kidney response in only a minority of affected individuals, and 10-15% of patients with LN develop kidney failure, with its attendant morbidity and considerable prognostic implications. Moreover, the medications most often used to treat LN - corticosteroids in combination with immunosuppressive or cytotoxic drugs - are associated with substantial side effects. Advances in proteomics, flow cytometry and RNA sequencing have led to important new insights into immune cells, molecules and mechanistic pathways that are instrumental in the pathogenesis of LN. These insights, together with a renewed focus on the study of human LN kidney tissue, suggest new therapeutic targets that are already being tested in lupus animal models and early-phase clinical trials and, as such, are hoped to eventually lead to meaningful improvements in the care of patients with systemic lupus erythematosus-associated kidney disease.
Collapse
Affiliation(s)
- Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| | - Ting Zhang
- Division of Rheumatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Division of Rheumatology and Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
12
|
Sundaresan B, Shirafkan F, Ripperger K, Rattay K. The Role of Viral Infections in the Onset of Autoimmune Diseases. Viruses 2023; 15:v15030782. [PMID: 36992490 PMCID: PMC10051805 DOI: 10.3390/v15030782] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Autoimmune diseases (AIDs) are the consequence of a breach in immune tolerance, leading to the inability to sufficiently differentiate between self and non-self. Immune reactions that are targeted towards self-antigens can ultimately lead to the destruction of the host's cells and the development of autoimmune diseases. Although autoimmune disorders are comparatively rare, the worldwide incidence and prevalence is increasing, and they have major adverse implications for mortality and morbidity. Genetic and environmental factors are thought to be the major factors contributing to the development of autoimmunity. Viral infections are one of the environmental triggers that can lead to autoimmunity. Current research suggests that several mechanisms, such as molecular mimicry, epitope spreading, and bystander activation, can cause viral-induced autoimmunity. Here we describe the latest insights into the pathomechanisms of viral-induced autoimmune diseases and discuss recent findings on COVID-19 infections and the development of AIDs.
Collapse
Affiliation(s)
- Bhargavi Sundaresan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Fatemeh Shirafkan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kevin Ripperger
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kristin Rattay
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
13
|
Zhang L. A common mechanism links Epstein-Barr virus infections and autoimmune diseases. J Med Virol 2023; 95:e28363. [PMID: 36451313 DOI: 10.1002/jmv.28363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Epstein-Barr virus (EBV) infection is associated with a variety of the autoimmune diseases. There is apparently no unified model for the role of EBV in autoimmune diseases. In this article, the development of autoimmune diseases is proposed as a simple two-step process: specific autoimmune initiators may cause irreversible changes to genetic materials that increase autoimmune risks, and autoimmune promoters promote autoimmune disease formation once cells are susceptible to autoimmunity. EBV has several types of latencies including type III latency with higher proliferation potential. EBV could serve as autoimmune initiators for some autoimmune diseases. At the same time, EBV may play a promotional role in majority of the autoimmune diseases by repeated replenishment of EBV type III latency cells and inflammatory cytokine productions in persistent stage. The type III latency cells have enhanced capacity as antigen-presenting cells that would facilitate the development of both B and T cell-mediated autoimmunity. The repeated cytokine productions are achieved by the repeated infection of naive B-lymphocytes and proliferation of type III latency cells that produce inflammatory cytokines. Presentation of viral or self-antigens by EBV type III latency B lymphocytes may promote autoreactive B cell and T cell proliferation, which can be amplified by type III latency cells-mediated cytokines productions. Different autoimmune diseases may require different kinds of pathogenic immune cells and/or specific cytokines. Frequency of the replenishment of EBV type III latency cells may determine the specific effect of the promoter functions. A specific initiator plus EBV-mediated common promoter function may lead to development of a specific autoimmune disease and link EBV-infection to a variety of autoimmunity.
Collapse
Affiliation(s)
- Luwen Zhang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska, Lincoln, Nebraska, USA
| |
Collapse
|
14
|
Vila IK, Guha S, Kalucka J, Olagnier D, Laguette N. Alternative pathways driven by STING: From innate immunity to lipid metabolism. Cytokine Growth Factor Rev 2022; 68:54-68. [PMID: 36085258 DOI: 10.1016/j.cytogfr.2022.08.006] [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: 08/21/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023]
Abstract
The Stimulator of Interferon Genes (STING) is a major adaptor protein that is central to the initiation of type I interferon responses and proinflammatory signalling. STING-dependent signalling is triggered by the presence of cytosolic nucleic acids that are generated following pathogen infection or cellular stress. Beyond this central role in controlling immune responses through the production of cytokines and chemokines, recent reports have uncovered inflammation-independent STING functions. Amongst these, a rapidly growing body of evidence demonstrates a key role of STING in controlling metabolic pathways at several levels. Since immunity and metabolic homeostasis are tightly interconnected, these findings deepen our understanding of the involvement of STING in human pathologies. Here, we discuss these findings and reflect on their impact on our current understanding of how nucleic acid immunity controls homeostasis and promotes pathological outcomes.
Collapse
Affiliation(s)
- Isabelle K Vila
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France.
| | - Soumyabrata Guha
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France
| | - Joanna Kalucka
- Aarhus University, Department of Biomedicine, Aarhus, Denmark
| | - David Olagnier
- Aarhus University, Department of Biomedicine, Aarhus, Denmark
| | - Nadine Laguette
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
15
|
Raftopoulou S, Rapti A, Karathanasis D, Evangelopoulos ME, Mavragani CP. The role of type I IFN in autoimmune and autoinflammatory diseases with CNS involvement. Front Neurol 2022; 13:1026449. [PMID: 36438941 PMCID: PMC9685560 DOI: 10.3389/fneur.2022.1026449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Type I interferons (IFNs) are major mediators of innate immunity, with well-known antiviral, antiproliferative, and immunomodulatory properties. A growing body of evidence suggests the involvement of type I IFNs in the pathogenesis of central nervous system (CNS) manifestations in the setting of chronic autoimmune and autoinflammatory disorders, while IFN-β has been for years, a well-established therapeutic modality for multiple sclerosis (MS). In the present review, we summarize the current evidence on the mechanisms of type I IFN production by CNS cellular populations as well as its local effects on the CNS. Additionally, the beneficial effects of IFN-β in the pathophysiology of MS are discussed, along with the contributory role of type I IFNs in the pathogenesis of neuropsychiatric lupus erythematosus and type I interferonopathies.
Collapse
Affiliation(s)
- Sylvia Raftopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Rapti
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Karathanasis
- First Department of Neurology, National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece
| | | | - Clio P. Mavragani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Meng J, Zhang J, Fang J, Li M, Ding H, Zhang W, Chen C. Dynamic inflammatory changes of the neurovascular units after ischemic stroke. Brain Res Bull 2022; 190:140-151. [DOI: 10.1016/j.brainresbull.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/21/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022]
|
17
|
Chen K, Wu T, Wang D, Li R, Shen X, Zhao T, Ozato K, Li R. Transcriptomics and quantitative proteomics reveal changes after second stimulation of bone marrow-derived macrophages from lupus-prone MRL/lpr mice. Front Immunol 2022; 13:1004232. [PMID: 36341359 PMCID: PMC9627492 DOI: 10.3389/fimmu.2022.1004232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immune memory can cause the occurrence and exacerbation of autoimmune diseases, and it is as well as being strongly associated with the pathogenesis of systemic lupus erythematosus (SLE), however, the specific mechanism remains to be further studied. We learned that IFN-γ stimulation generated innate immune memory in bone marrow-derived macrophages (BMDMs) and activated memory interferon-stimulated genes (ISGs). This research used IFN-γ and lipopolysaccharide (LPS) to treat BMDMs with lupus-prone MRL/lpr mice and showed that particular memory ISGs were substantially elevated in prestimulated macrophages. In order to identify the differentially expressed genes (DEGs), researchers turned to RNA-seq. GO and KEGG analysis showed that up-regulated DEGs were enriched in defense and innate immune responses, and were related to the expression of pattern recognition receptors (PRRs)-related pathways in macrophages. TMT-based proteome analysis revealed differentially expressed proteins (DEPs) up-regulated in BMDMs were abundant in metabolic pathways such as glucose metabolism. Our study found that after the secondary stimulation of MRL/lpr mice, the expression of PRRs in innate immune cells was changed, and IFN-related pathways were activated to release a large number of ISGs to promote the secondary response. At the same time, related metabolic modes such as glycolysis were enhanced, and epigenetic changes may occur. Therefore, SLE is brought on, maintained, and worsened by a variety of factors that work together to produce innate immune memory.
Collapse
Affiliation(s)
- Keyue Chen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tiyun Wu
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Danyan Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rong Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangfeng Shen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Keiko Ozato, ; Rongqun Li,
| | - Rongqun Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Keiko Ozato, ; Rongqun Li,
| |
Collapse
|
18
|
Itotagawa E, Tomofuji Y, Kato Y, Konaka H, Tsujimoto K, Park J, Nagira D, Hirayama T, Jo T, Hirano T, Morita T, Nishide M, Nishida S, Shima Y, Narazaki M, Okada Y, Takamatsu H, Kumanogoh A. SLE stratification based on BAFF and IFN-I bioactivity for biologics and implications of BAFF produced by glomeruli in lupus nephritis. Rheumatology (Oxford) 2022; 62:1988-1997. [PMID: 36094336 PMCID: PMC10152287 DOI: 10.1093/rheumatology/keac528] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/30/2022] [Accepted: 09/03/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE B-cell activating factor (BAFF) is implicated in systemic lupus erythematosus (SLE) pathogenesis. Blocking BAFF signaling has contributed to reducing glucocorticoid dosage and preventing organ damage. However, clinical characteristics of patients who may benefit from this therapy are not yet fully elucidated. Therefore, we identified patients with high BAFF-bioactivity to investigate their clinical characteristics and BAFF-producing cells. METHODS We established the reporter cell for BAFF and investigated the clinical characteristics of SLE patients with high BAFF-bioactivity. We identified BAFF-expressing kidney cells using publicly available scRNA-seq data and immunohistological analysis. SLE patients were stratified based on the bioactivity of BAFF and type-I interferon (IFN-I) to identify associated characteristic clinical manifestations. RESULTS SLE patients, especially patients with lupus nephritis (LN), had significantly higher serum BAFF-bioactivity than healthy controls (HC) and non-LN patients. Additionally, single-cell-RNA-seq data and immunohistological analysis of kidney samples from LN patients revealed that BAFF is expressed in glomerular macrophages and mesangial cells. Notably, BAFF bioactivity was elevated in the urine of LN patients compared to that of non-LN patients, while no IFN-I bioactivity was detected in the urine. Furthermore, SLE stratification based on bioactivities of serum BAFF and IFN-I revealed the clinical characteristics of patients: high BAFF represented patients with LN and high IFN-I represented patients with blood and skin manifestations. CONCLUSIONS Monitoring urinary BAFF-bioactivity may be valuable in diagnosing LN. Furthermore, stratification based on serum BAFF and IFN-I bioactivities may allow the identification of appropriate patients for biologics targeting BAFF and IFN-I.
Collapse
Affiliation(s)
- Eri Itotagawa
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihiko Tomofuji
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita Osaka, 565-0871, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hachiro Konaka
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of internal medicine, Nippon life Hospital, 2-1-54 Enokojima, Nishi-ku, Osaka, Osaka 550-0006, Japan
| | - Kohei Tsujimoto
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - JeongHoon Park
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of internal medicine, Daini Osaka Police Hospital, Osaka, 543-8922, Japan
| | - Daiki Nagira
- Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takehiro Hirayama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tatsunori Jo
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toru Hirano
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Rheumatology, Nishinomiya municipal central hospital, 8-24 Hayashida-cho, Nishinomiya, Hyogo, 663-8014, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Division of Thermo-Therapeutics for Vascular Dysfunction, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita Osaka, 565-0871, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan.,Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi 230-0045, Japan.,Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Immunopathology, WPI, Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| |
Collapse
|
19
|
Giordano AMS, Luciani M, Gatto F, Abou Alezz M, Beghè C, Della Volpe L, Migliara A, Valsoni S, Genua M, Dzieciatkowska M, Frati G, Tahraoui-Bories J, Giliani SC, Orcesi S, Fazzi E, Ostuni R, D'Alessandro A, Di Micco R, Merelli I, Lombardo A, Reijns MAM, Gromak N, Gritti A, Kajaste-Rudnitski A. DNA damage contributes to neurotoxic inflammation in Aicardi-Goutières syndrome astrocytes. J Exp Med 2022; 219:213058. [PMID: 35262626 PMCID: PMC8916121 DOI: 10.1084/jem.20211121] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 11/22/2021] [Accepted: 01/24/2022] [Indexed: 01/09/2023] Open
Abstract
Aberrant induction of type I IFN is a hallmark of the inherited encephalopathy Aicardi-Goutières syndrome (AGS), but the mechanisms triggering disease in the human central nervous system (CNS) remain elusive. Here, we generated human models of AGS using genetically modified and patient-derived pluripotent stem cells harboring TREX1 or RNASEH2B loss-of-function alleles. Genome-wide transcriptomic analysis reveals that spontaneous proinflammatory activation in AGS astrocytes initiates signaling cascades impacting multiple CNS cell subsets analyzed at the single-cell level. We identify accumulating DNA damage, with elevated R-loop and micronuclei formation, as a driver of STING- and NLRP3-related inflammatory responses leading to the secretion of neurotoxic mediators. Importantly, pharmacological inhibition of proapoptotic or inflammatory cascades in AGS astrocytes prevents neurotoxicity without apparent impact on their increased type I IFN responses. Together, our work identifies DNA damage as a major driver of neurotoxic inflammation in AGS astrocytes, suggests a role for AGS gene products in R-loop homeostasis, and identifies common denominators of disease that can be targeted to prevent astrocyte-mediated neurotoxicity in AGS.
Collapse
Affiliation(s)
- Anna Maria Sole Giordano
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Francesca Gatto
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Monah Abou Alezz
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Beghè
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Lucrezia Della Volpe
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Alessandro Migliara
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Sara Valsoni
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Marco Genua
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Giacomo Frati
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Julie Tahraoui-Bories
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Clara Giliani
- Department of Molecular and Translational Medicine, "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Azienda Socio Sanitaria Territoriale Spedali Civili, Brescia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Child Neurology and Psychiatry Unit, Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia, Italy
| | - Elisa Fazzi
- Unit of Child Neurology and Psychiatry, Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Azienda Socio Sanitaria Territoriale Spedali Civili, Brescia, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Martin A M Reijns
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Natalia Gromak
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
20
|
Hasse S, Julien AS, Duchez AC, Zhao C, Boilard E, Fortin PR, Bourgoin SG. Red blood cell-derived phosphatidylserine positive extracellular vesicles are associated with past thrombotic events in patients with systemic erythematous lupus. Lupus Sci Med 2022; 9:9/1/e000605. [PMID: 35260475 PMCID: PMC8905995 DOI: 10.1136/lupus-2021-000605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022]
Abstract
Background Extracellular vesicles (EVs) released by blood cells have proinflammation and procoagulant action. Patients with systemic lupus erythematosus (SLE) present high vascular inflammation and are prone to develop cardiovascular diseases. Therefore, we postulated that the EV populations found in blood, including platelet EVs (PEVs) and red blood cell EVs (REVs), are associated with SLE disease activity and SLE-associated cardiovascular accidents. Method We assessed autotaxin (ATX) plasma levels by ELISA, the platelet activation markers PAC1 and CD62P, ATX bound to platelets and the amounts of plasma PEVs and REVs by flow cytometry in a cohort of 102 patients with SLE, including 29 incident cases of SLE and 30 controls. Correlation analyses explored the associations with the clinical parameters. Result Platelet activation markers were increased in patients with SLE compared with healthy control, with the marker CD62P associated with the SLE disease activity index (SLEDAI). The incident cases show additional associations between platelet markers (CD62P/ATX and PAC1/CD62P) and the SLEDAI. Compared with controls, patients with SLE presented higher levels of PEVs, phosphatidylserine positive (PS+) PEVs, REVs and PS+ REVs, but there is no association with disease activity. When stratified according to the plasma level of PS+ REVs, the group of patients with SLE with a high level of PS+ REVs presented a higher number of past thrombosis events and higher ATX levels. Conclusion Incident and prevalent forms of SLE cases present similar levels of platelet activation markers, with CD62P correlating with disease activity. Though EVs are not associated with disease activity, the incidence of past thrombotic events is higher in patients with a high level of PS+ REVs.
Collapse
Affiliation(s)
- Stephan Hasse
- Axe Maladies Infectieuses et Immunitaires, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistique, Université Laval, Quebec city, Quebec, Canada
| | - Anne-Claire Duchez
- Axe Maladies Infectieuses et Immunitaires, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| | - Chenqi Zhao
- Axe Maladies Infectieuses et Immunitaires, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| | - Eric Boilard
- Département de microbiologie-infectiologie et immunologie, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| | - Paul R Fortin
- Département de Médecine, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| | - Sylvain G Bourgoin
- Département de microbiologie-infectiologie et immunologie, Centre de recherche du CHU de Québec-Université Laval, Centre ARThrite de l'Université Laval, Quebec city, Quebec, Canada
| |
Collapse
|
21
|
Skudalski L, Shahriari N, Torre K, Santiago S, Bibb L, Kodomudi V, Grant-Kels JM, Lu J. Emerging Therapeutics in the Management of Connective Tissue Disease. Part I. Lupus Erythematosus and Sjögren's Syndrome. J Am Acad Dermatol 2022; 87:1-18. [PMID: 35202775 DOI: 10.1016/j.jaad.2021.12.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022]
Abstract
The management of connective tissue diseases is dramatically evolving with the advent of biologics and novel oral systemic therapeutics. Despite involvement in the care of these complex patients, there is a knowledge gap in the field of dermatology regarding these emerging agents. The first article in this continuing medical education series discusses new and emerging therapeutics for lupus erythematosus and Sjögren's syndrome that target cells, intracellular signaling pathways, and cytokines.
Collapse
Affiliation(s)
| | - Neda Shahriari
- Department of Dermatology, Brigham and Women's Hospital; Harvard Medical School, Boston, MA
| | - Kristin Torre
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT
| | - Sueheidi Santiago
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT
| | - Lorin Bibb
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT
| | - Vijay Kodomudi
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT
| | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT
| | - Jun Lu
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT.
| |
Collapse
|
22
|
Guo G, Wang H, Tong X, Ye L, Shi X, Fang S, Hu Y, Han F, Chen C, Ding N, Su B, Xue X, Zhang H. Transcriptional Landscape of Enhancer RNAs in Peripheral Blood Mononuclear Cells from Patients with Systemic Lupus Erythematosus. J Inflamm Res 2022; 15:775-791. [PMID: 35153501 PMCID: PMC8824297 DOI: 10.2147/jir.s331188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/11/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Enhancer RNAs (eRNAs), a class of non-coding RNAs, play indispensable roles in regulating target gene transcription and maintaining cell identity in cooperation with promoters. In this study, we investigated the transcriptional landscape and potential functions of eRNAs in peripheral blood mononuclear cells (PBMCs) from patients with systemic lupus erythematosus (SLE). Methods PBMCs from five patients with stable SLE, five patients with active SLE, and ten healthy individuals (HCs) were subjected to RNA-sequencing. Putative regulators, differential expression, and pathways were analyzed. eRNAs that were significantly upregulated were first validated by RT-qPCR in 12 samples. Then, candidate eRNAs were confirmed in a validation cohort of 45 samples. We conducted comprehensive pathway analyses to explore the correlations between the candidate eRNAs and SLE pathology. Results By analyzing eRNA transcript data from PBMCs from SLE patients and HCs, we identified various eRNAs and functional super-enhancers potentially related with SLE. The SLE-specificity of eRNAs seemed to be largely driven by SLE-specific transcription factors (TFs). A Venn diagram of eRNAs differentially expressed in stable, active, and total SLE vs HCs revealed that 13 and 23 eRNAs were commonly upregulated and downregulated, respectively, in patients with stable SLE and those with active SLE. The commonly upregulated eRNAs participate in regulating SLE-related pathways. Only eRNA TCONS_00034326 was significantly (P < 0.05) upregulated in PBMCs of patients with SLE when compared with those of HCs as indicated by RT-qPCR. The area under the receiver-operating curve of TCONS_00034326 for distinguishing SLE patients from HCs was 0.691. Through its putative SLE-related master TF, TCONS_00034326 is involved in multiple SLE-relevant signaling pathways, especially tumor necrosis factor signaling. Conclusion This study unraveled the transcriptional landscape of eRNAs, eRNA-related TFs, and super-enhancers in PBMCs from SLE patients and HCs. We identified a panel of SLE-relevant eRNAs, providing potential targets in SLE pathogenesis.
Collapse
Affiliation(s)
- Gangqiang Guo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Huijing Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, People’s Republic of China
| | - Xinya Tong
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Lele Ye
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Xinyu Shi
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Su Fang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Ya Hu
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, People’s Republic of China
| | - Chaosheng Chen
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Ning Ding
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Bofeng Su
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
- Correspondence: Bofeng Su; Huidi Zhang, Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China, Email ;
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens & Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Huidi Zhang
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| |
Collapse
|
23
|
Iwamoto T, Dorschner JM, Selvaraj S, Mezzano V, Jensen MA, Vsetecka D, Amin S, Makol A, Osborn T, Moder K, Chowdhary VR, Izmirly P, Belmont HM, Clancy RM, Buyon JP, Wu M, Loomis CA, Niewold TB. High Systemic Type I Interferon Activity is Associated with Active Class III/IV Lupus Nephritis. J Rheumatol 2021; 49:388-397. [PMID: 34782453 DOI: 10.3899/jrheum.210391] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Previous studies suggest a link between high serum type I interferon (IFN) and lupus nephritis (LN). We determined whether serum IFN activity is associated with subtypes of LN and studied renal tissues and cells to understand the impact of IFN in LN. METHODS 221 systemic lupus erythematosus (SLE) patients were studied. Serum IFN activity was measured by WISH bioassay. mRNA in-situ hybridization was used in renal tissue to measure expression of the representative IFN-induced gene, interferon-induced protein with tetratricopeptide repeats-1 (IFIT1), and the plasmacytoid dendritic cell (pDC) marker gene C-type lectin domain family-4 member C (CLEC4C or BDCA2). Podocyte cell line gene expression was measured by real-time PCR. RESULTS Class III/IV LN prevalence was significantly increased in patients with high serum IFN compared with those with low IFN (OR=5.48, p=4.0x10-7). In multivariate regression models, type I IFN was a stronger predictor of class III/IV LN than complement C3 or anti-dsDNA antibody, and could account for the association of these variables with LN. IFIT1 expression was increased in all classes of LN, but most in the glomerular areas of active class III/IV LN kidneys. IFIT1 expression was not closely co-localized with pDCs. IFN directly activated podocyte cell lines to induce chemokines and proapoptotic molecules. CONCLUSION Systemic high IFN is involved in the pathogenesis of severe LN. We do not find co-localization of pDCs with IFN signature in renal tissue, and instead observe the greatest intensity of IFN signature in glomerular areas, which could suggest a blood source of IFN.
Collapse
Affiliation(s)
- Taro Iwamoto
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Jessica M Dorschner
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Shanmugapriya Selvaraj
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Valeria Mezzano
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Mark A Jensen
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Danielle Vsetecka
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Shreyasee Amin
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Ashima Makol
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Thomas Osborn
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Kevin Moder
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Vaidehi R Chowdhary
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Peter Izmirly
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - H Michael Belmont
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Robert M Clancy
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Jill P Buyon
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Ming Wu
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Cynthia A Loomis
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| | - Timothy B Niewold
- Colton Center for Autoimmunity, New York University, USA; Allergy and Clinical Immunology, Chiba University, Japan; Mayo Clinic College of Medicine, USA; Department of Pathology, New York University, USA; Division of Rheumatology, New York University, USA. Funding: TBN: Grants from the Colton Center for Autoimmunity, NIH (AR060861, AR057781, AR065964), the Lupus Research Foundation, and the Lupus Research Alliance Disclosures of Competing Interests: TBN has received research grants from EMD Serono and Janssen, and has consulted for Thermo Fisher, Progentec, and Inova, all unrelated to the current manuscript. Corresponding author: Timothy B. Niewold, MD, Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016,
| |
Collapse
|
24
|
Tan PH, Ji J, Yeh CC, Ji RR. Interferons in Pain and Infections: Emerging Roles in Neuro-Immune and Neuro-Glial Interactions. Front Immunol 2021; 12:783725. [PMID: 34804074 PMCID: PMC8602180 DOI: 10.3389/fimmu.2021.783725] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Interferons (IFNs) are cytokines that possess antiviral, antiproliferative, and immunomodulatory actions. IFN-α and IFN-β are two major family members of type-I IFNs and are used to treat diseases, including hepatitis and multiple sclerosis. Emerging evidence suggests that type-I IFN receptors (IFNARs) are also expressed by microglia, astrocytes, and neurons in the central and peripheral nervous systems. Apart from canonical transcriptional regulations, IFN-α and IFN-β can rapidly suppress neuronal activity and synaptic transmission via non-genomic regulation, leading to potent analgesia. IFN-γ is the only member of the type-II IFN family and induces central sensitization and microglia activation in persistent pain. We discuss how type-I and type-II IFNs regulate pain and infection via neuro-immune modulations, with special focus on neuroinflammation and neuro-glial interactions. We also highlight distinct roles of type-I IFNs in the peripheral and central nervous system. Insights into IFN signaling in nociceptors and their distinct actions in physiological vs. pathological and acute vs. chronic conditions will improve our treatments of pain after surgeries, traumas, and infections.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts, MA, United States
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Chun-Chang Yeh
- Department of Anesthesiology of Tri-Service General Hospital & National Defense Medical Center, Taipei City, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
- Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
25
|
Musella M, Galassi C, Manduca N, Sistigu A. The Yin and Yang of Type I IFNs in Cancer Promotion and Immune Activation. BIOLOGY 2021; 10:856. [PMID: 34571733 PMCID: PMC8467547 DOI: 10.3390/biology10090856] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/22/2022]
Abstract
Type I Interferons (IFNs) are key regulators of natural and therapy-induced host defense against viral infection and cancer. Several years of remarkable progress in the field of oncoimmunology have revealed the dual nature of these cytokines. Hence, Type I IFNs may trigger anti-tumoral responses, while leading immune dysfunction and disease progression. This dichotomy relies on the duration and intensity of the transduced signaling, the nature of the unleashed IFN stimulated genes, and the subset of responding cells. Here, we discuss the role of Type I IFNs in the evolving relationship between the host immune system and cancer, as we offer a view of the therapeutic strategies that exploit and require an intact Type I IFN signaling, and the role of these cytokines in inducing adaptive resistance. A deep understanding of the complex, yet highly regulated, network of Type I IFN triggered molecular pathways will help find a timely and immune"logical" way to exploit these cytokines for anticancer therapy.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Claudia Galassi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (C.G.); (N.M.)
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
26
|
Osman EMA, Abu El Nazar SY, Maharem DA, Al-Jebouri DM, Naga IS. Relation between Vitamin D Level and Cyclin-Dependent Kinase-1 Gene Expression in Egyptian Patients with Lupus Nephritis and their Impact on Disease Activity. Indian J Nephrol 2021; 31:163-168. [PMID: 34267439 PMCID: PMC8240929 DOI: 10.4103/ijn.ijn_359_19] [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: 10/28/2019] [Revised: 02/04/2020] [Accepted: 03/08/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction: Lupus nephritis (LN) is a common complication of systemic lupus erythematosus. Vitamin D and cycline-dependent kinase-1 (CDK1) have been implicated in its pathogenesis. The aim of this study was to determine the relation between vitamin D level and CDK-1 in lupus nephritis patients and their impact on disease activity. Patients and Methods: The current study was conducted on 50 LN patients and 20 control subjects from Egyptian population using ELISA to assess vitamin D level in serum and TaqMan assay for CDK1 gene expression. Results: Serum vitamin D level was significantly lower in LN patients and CDK-1 gene was down expressed in the majority of LN patients. A significant inverse correlation was found between vitamin D level and 24 h protein in urine, ANA, anti-dsDNA, CRP, with a significant positive correlation with renal biopsy indices, eGFR. There was a non-significant inverse correlation between vitamin D and CDK-1 (before RO-3306 addition) and a positive correlation after RO-3306. A significant positive correlation was found between CDK-1 gene expressions with urinary albumin/creatinine ratio. However, a significant positive correlation was found between CDK-1 (after RO-3306 addition) and proteinuria. While a significant positive correlation was found between CDK-1 expression (after RO-3306 addition) and ANA, a significant positive correlation was found between CDK-1 expression (before RO-3306 addition) and anti-dsDNA but CDK-1 is not associated with renal biopsy indices nor with activity indices of SLE. There was a positive correlation between CDK-1 gene expression and CRP before and after RO-3306 addition. Conclusions: Vitamin D deficiency acts as a risk factor for developing LN. CDK-1 may have an association with the diagnosis of LN but its association with the progression of staging of LN is still confusing
Collapse
Affiliation(s)
- Eman M A Osman
- Department of Immunology and Allergy Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Salma Y Abu El Nazar
- Department of Immunology and Allergy Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Dalia A Maharem
- Internal Medicine, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Iman S Naga
- Microbiology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
27
|
Brawerman G, Thompson PJ. Beta Cell Therapies for Preventing Type 1 Diabetes: From Bench to Bedside. Biomolecules 2020; 10:E1681. [PMID: 33339173 PMCID: PMC7765619 DOI: 10.3390/biom10121681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic metabolic disease characterized by insulin deficiency, generally resulting from progressive autoimmune-mediated destruction of pancreatic beta cells. While the phenomenon of beta cell autoimmunity continues to be an active area of investigation, recent evidence suggests that beta cell stress responses are also important contributors to disease onset. Here we review the pathways driving different kinds of beta cell dysfunction and their respective therapeutic targets in the prevention of T1D. We discuss opportunities and important open questions around the effectiveness of beta cell therapies and challenges for clinical utility. We further evaluate ways in which beta cell drug therapy could be combined with immunotherapy for preventing T1D in light of our growing appreciation of disease heterogeneity and patient endotypes. Ultimately, the emergence of pharmacologic beta cell therapies for T1D have armed us with new tools and closing the knowledge gaps in T1D etiology will be essential for maximizing the potential of these approaches.
Collapse
Affiliation(s)
- Gabriel Brawerman
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Peter J. Thompson
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
28
|
Ma Z, Zhang W, Fan W, Wu Y, Zhang M, Xu J, Li W, Sun L, Liu W, Liu W. Forkhead box O1-mediated ubiquitination suppresses RIG-I-mediated antiviral immune responses. Int Immunopharmacol 2020; 90:107152. [PMID: 33187908 DOI: 10.1016/j.intimp.2020.107152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022]
Abstract
RNA virus infection activates the RIG-I-like Receptor (RLR) signaling pathway to produce type I interferons (IFNs), the key components of the antiviral immune response. Forkhead box O1 (FoxO1) is a host transcription factor that participates in multiple biological processes. In this study, FoxO1 was identified as a critical negative regulator of RIG-I-triggered signaling. FoxO1 promoted Sendai virus (SeV) replication and downregulated type I IFN production. Upon SeV infection, FoxO1 suppressed K63-linked ubiquitination of TRAF3 and the interaction between TRAF3 and TBK1, after which the production of type I IFNs via the interferon regulatory transcription factor 3 (IRF3) pathways was reduced. In addition, FoxO1 destabilized IRF3 by facilitating E3 ligase TRIM22- or TRIM21-mediated K48-linked ubiquitination of IRF3. Moreover, the inhibitory effect of FoxO1 was found to depend on its DNA binding domain (DBD). Thus, our findings highlight novel important roles of FoxO1 in controlling RLR-mediated antiviral innate immunity.
Collapse
Affiliation(s)
- Zhenling Ma
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenwen Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenhui Fan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yaru Wu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Menghao Zhang
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Jun Xu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenqing Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China
| | - Lei Sun
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China.
| |
Collapse
|
29
|
Kang JA, Jeon YJ. Emerging Roles of USP18: From Biology to Pathophysiology. Int J Mol Sci 2020; 21:ijms21186825. [PMID: 32957626 PMCID: PMC7555095 DOI: 10.3390/ijms21186825] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Eukaryotic proteomes are enormously sophisticated through versatile post-translational modifications (PTMs) of proteins. A large variety of code generated via PTMs of proteins by ubiquitin (ubiquitination) and ubiquitin-like proteins (Ubls), such as interferon (IFN)-stimulated gene 15 (ISG15), small ubiquitin-related modifier (SUMO) and neural precursor cell expressed, developmentally downregulated 8 (NEDD8), not only provides distinct signals but also orchestrates a plethora of biological processes, thereby underscoring the necessity for sophisticated and fine-tuned mechanisms of code regulation. Deubiquitinases (DUBs) play a pivotal role in the disassembly of the complex code and removal of the signal. Ubiquitin-specific protease 18 (USP18), originally referred to as UBP43, is a major DUB that reverses the PTM of target proteins by ISG15 (ISGylation). Intriguingly, USP18 is a multifaceted protein that not only removes ISG15 or ubiquitin from conjugated proteins in a deconjugating activity-dependent manner but also acts as a negative modulator of type I IFN signaling, irrespective of its catalytic activity. The function of USP18 has become gradually clear, but not yet been completely addressed. In this review, we summarize recent advances in our understanding of the multifaceted roles of USP18. We also highlight new insights into how USP18 is implicated not only in physiology but also in pathogenesis of various human diseases, involving infectious diseases, neurological disorders, and cancers. Eventually, we integrate a discussion of the potential of therapeutic interventions for targeting USP18 for disease treatment.
Collapse
Affiliation(s)
- Ji An Kang
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Young Joo Jeon
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon 35015, Korea;
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Correspondence: ; Tel.: +82-42-280-6766; Fax: +82-42-280-6769
| |
Collapse
|
30
|
Nocito C, Lubinsky C, Hand M, Khan S, Patel T, Seliga A, Winfield M, Zuluaga-Ramirez V, Fernandes N, Shi X, Unterwald EM, Persidsky Y, Sriram U. Centrally Acting Angiotensin-Converting Enzyme Inhibitor Suppresses Type I Interferon Responses and Decreases Inflammation in the Periphery and the CNS in Lupus-Prone Mice. Front Immunol 2020; 11:573677. [PMID: 33042154 PMCID: PMC7522287 DOI: 10.3389/fimmu.2020.573677] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multi-organ damage. Neuropsychiatric lupus (NPSLE) is one of the most common manifestations of human SLE, often causing depression. Interferon-α (IFNα) is a central mediator in disease pathogenesis. Administration of IFNα to patients with chronic viral infections or cancers causes depressive symptoms. Angiotensin-converting enzyme (ACE) is part of the kallikrein-kinin/renin-angiotensin (KKS/RAS) system that regulates many physiological processes, including inflammation, and brain functions. It is known that ACE degrades bradykinin (BK) into inactive peptides. We have previously shown in an in vitro model of mouse bone-marrow-derived dendritic cells (BMDC) and human peripheral blood mononuclear cells that captopril (a centrally acting ACE inhibitor-ACEi) suppressed Type I IFN responsive gene (IRG) expression. In this report, we used the MRL/lpr lupus-prone mouse model, an established model to study NPSLE, to determine the in vivo effects of captopril on Type I IFN and associated immune responses in the periphery and brain and effects on behavior. Administering captopril to MRL/lpr mice decreased expression of IRGs in brain, spleen and kidney, decreased circulating and tissue IFNα levels, decreased microglial activation (IBA-1 expression) and reduced depressive-like behavior. Serotonin levels that are decreased in depression were increased by captopril treatment. Captopril also reduced autoantibody levels in plasma and immune complex deposition in kidney and brain. Thus, ACEi's may have potential for therapeutic use for systemic and NPSLE.
Collapse
Affiliation(s)
- Cassandra Nocito
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Cody Lubinsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Michelle Hand
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Sabeeya Khan
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Tulsi Patel
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Alecia Seliga
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Viviana Zuluaga-Ramirez
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Nicole Fernandes
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Xiangdang Shi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
31
|
Cai C, Yu X. A mathematic model to reveal delicate cross-regulation between MAVS/STING, inflammasome and MyD88-dependent type I interferon signalling. J Cell Mol Med 2020; 24:11535-11545. [PMID: 32885594 PMCID: PMC7576308 DOI: 10.1111/jcmm.15768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Early type I interferon is essential for antagonizing against malaria infection, which remains a significant global infectious disease. After Plasmodium yoelii YM infection, the activation of MAVS‐, STING‐ and inflammasome‐IRF3‐mediated pathway could trigger the Socs1 expression to inhibit the TLR7‐MyD88‐IRF7‐induced type I interferon production. However, the dynamic regulatory mechanisms of type I interferon response to YM infection and delicate cross‐regulation of these signalling are far from clear. In current study, we established a mathematical model to systematically demonstrate that the MAVS‐, STING‐ and inflammasome‐mediated signalling pathways play distinct roles in regulating type I interferon response after YM infection; and the YM dose could significantly affect the difference of resistance to YM infection among MAVS, STING and inflammasome deficiency. Collectively, our study systematically elucidated the precise regulatory mechanisms of type I interferon signalling after YM infection and advanced the research on therapy of plasmodium infection by incorporating multiple signalling pathways at diverse time.
Collapse
Affiliation(s)
- Chunmei Cai
- Research Center for High Altitude Medicine, School of Medical, Qinghai University, Xining, China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Xining, China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Hiepe F. Neue Erkenntnisse zur Pathogenese des SLE und ihre Auswirkungen auf
die Entwicklung neuer Therapie-Konzepte. AKTUEL RHEUMATOL 2020. [DOI: 10.1055/a-1210-2259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
ZusammenfassungAutoantikörper sind essentiell in der Pathogenese des SLE. Sie sind das
Ergebnis einer Störung des erworbenen (adaptiven) Immunsystems mit
fehlender Toleranz gegen Selbst. Eine Typ-I Interferon-Signatur, die im
angeborenen (innaten) Immunsystem ihren Ursprung hat, ist ein wesentlicher
Treiber dieser Störung. Autoantikörper können sowohl von
kurzlebigen, proliferierenden Plasmablasten, die B-Zell-Hyperaktivität
widerspiegeln, als auch von langlebigen, nicht-proliferierenden
Gedächtnis-Plasmazellen sezerniert werden.
Gedächtnis-Plasmazellen, die in Nischen im Knochenmark und im
entzündeten Gewebe lokalisiert sind, lassen sich nicht durch
konventionelle Immunsuppressiva und Therapien mit B-Zellen als Target
eliminieren. Konzepte, die auf die Depletion von Gedächtnis-Plasmazellen
abzielen, können im Zusammenspiel mit Targets, die eine Aktivierung von
autoreaktiven B-Zellen verhindern, ein kuratives Potenzial haben.
Collapse
Affiliation(s)
- Falk Hiepe
- Medizinische Klinik mit Schwerpunkt Rheumatologie und klin.
Immunologie, Charité – Universitätsmedizin Berlin;
Deutsches Rheumaforschungszentrum – ein Institut der
Leibniz-Gemeinschaft, Berlin
| |
Collapse
|
33
|
Katsuyama T, Li H, Comte D, Tsokos GC, Moulton VR. Splicing factor SRSF1 controls T cell hyperactivity and systemic autoimmunity. J Clin Invest 2020; 129:5411-5423. [PMID: 31487268 DOI: 10.1172/jci127949] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 09/03/2019] [Indexed: 01/25/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a devastating autoimmune disease in which hyperactive T cells play a critical role. Understanding molecular mechanisms underlying the T cell hyperactivity will lead to identification of specific therapeutic targets. Serine/arginine-rich splicing factor 1 (SRSF1) is an essential RNA-binding protein that controls posttranscriptional gene expression. We have demonstrated that SRSF1 levels are aberrantly decreased in T cells from patients with SLE and that they correlate with severe disease, yet the role of SRSF1 in T cell physiology and autoimmune disease is largely unknown. Here we show that T cell-restricted Srsf1-deficient mice develop systemic autoimmunity and lupus-nephritis. Mice exhibit increased frequencies of activated/effector T cells producing proinflammatory cytokines, and an elevated T cell activation gene signature. Mechanistically, we noted increased activity of the mechanistic target of rapamycin (mTOR) pathway and reduced expression of its repressor PTEN. The mTOR complex 1 (mTORC1) inhibitor rapamycin suppressed proinflammatory cytokine production by T cells and alleviated autoimmunity in Srsf1-deficient mice. Of direct clinical relevance, PTEN levels correlated with SRSF1 in T cells from patients with SLE, and SRSF1 overexpression rescued PTEN and suppressed mTORC1 activation and proinflammatory cytokine production. Our studies reveal the role of a previously unrecognized molecule, SRSF1, in restraining T cell activation, averting the development of autoimmune disease, and acting as a potential therapeutic target for lupus.
Collapse
Affiliation(s)
- Takayuki Katsuyama
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Hao Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Denis Comte
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Divisions of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Abstract
Innate lymphocyte populations are emerging as key effectors in tissue homeostasis, microbial defense, and inflammatory skin disease. The cells are evolutionarily ancient and carry conserved principles of function, which can be achieved through shared or unique specific mechanisms. Recent technological and treatment advances have provided insight into heterogeneity within and between individuals and species. Similar pathways can extend through to adaptive lymphocytes, which softens the margins with innate lymphocyte populations and allows investigation of nonredundant pathways of immunity and inflammation that might be amenable to therapeutic intervention. Here, we review advances in understanding of innate lymphocyte biology with a focus on skin disease and the roles of commensal and pathogen responses and tissue homeostasis.
Collapse
Affiliation(s)
- Yi-Ling Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Clare S Hardman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Koshika Yadava
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Graham Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals, Headington, Oxford OX3 7LE, United Kingdom;
| |
Collapse
|
35
|
Rubin LH, Xu Y, Norris PJ, Wang X, Dastgheyb R, Fitzgerald KC, Keating SM, Kaplan RC, Maki PM, Anastos K, Springer G, Benning L, Kassaye S, Gustafson DR, Valcour VG, Williams DW. Early Inflammatory Signatures Predict Subsequent Cognition in Long-Term Virally Suppressed Women With HIV. Front Integr Neurosci 2020; 14:20. [PMID: 32390808 PMCID: PMC7193823 DOI: 10.3389/fnint.2020.00020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022] Open
Abstract
Immunologic function is an important determinant of cognition. Here we examined the contribution of early immune signatures to cognitive performance among HIV-infected, virally suppressed women (HIV+VS) and in HIV-uninfected (HIV-) women. Specifically, we measured serum inflammatory markers, developed combinatory immune signatures, and evaluated their associations with cognition. Forty-nine HIV+VS women in the Women’s Interagency HIV Study (WIHS) who achieved viral suppression shortly after effective antiretroviral therapy (ART) initiation, and 56 matched HIV− women were selected. Forty-two serum inflammatory markers were measured within 2 years of effective ART initiation for HIV+VS women, and at an initial timepoint for HIV− women. The same inflammatory markers were also measured approximately 1, 7, and 12 years later for all women. Of the 105 women with complete immune data, 83 (34 HIV+VS, 49 HIV−) also had cognitive data available 12 years later at ≥1 time points (median = 3.1). We searched for combinatory immune signatures by adapting a dynamic matrix factorization analytic method that builds upon Tucker decomposition followed by Ingenuity® Pathway Analysis to facilitate data interpretation. Seven combinatory immune signatures emerged based on the Frobenius residual. Three signatures were common between HIV+VS and HIV− women, while four signatures were unique. These inflammatory signatures predicted subsequent cognitive performance in both groups using mixed-effects modeling, but more domain-specific associations were significant in HIV+VS than HIV− women. Leukocyte influx into brain was a major contributor to cognitive function in HIV+VS women, while T cell exhaustion, inflammatory response indicative of depressive/psychiatric disorders, microglial activity, and cytokine signaling predicted both global and domain-specific performance for HIV− women. Our findings suggest that immune signatures may be useful diagnostic, prognostic, and immunotherapeutic targets predictive of subsequent cognitive performance. Importantly, they also provide insight into common and distinct inflammatory mechanisms underlying cognition in HIV− and HIV+VS women.
Collapse
Affiliation(s)
- Leah H Rubin
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States.,Department of Psychiatry, Johns Hopkins University, Baltimore, MD, United States.,Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Philip J Norris
- Department of Laboratory Medicine, Vitalant Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Xuzhi Wang
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States
| | - Raha Dastgheyb
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | | | - Sheila M Keating
- Department of Laboratory Medicine, Vitalant Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States.,Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of General Internal Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gayle Springer
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lorie Benning
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Seble Kassaye
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Deborah R Gustafson
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Victor G Valcour
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Dionna W Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, United States.,Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
Shirley JL, Keeler GD, Sherman A, Zolotukhin I, Markusic DM, Hoffman BE, Morel LM, Wallet MA, Terhorst C, Herzog RW. Type I IFN Sensing by cDCs and CD4 + T Cell Help Are Both Requisite for Cross-Priming of AAV Capsid-Specific CD8 + T Cells. Mol Ther 2020; 28:758-770. [PMID: 31780366 PMCID: PMC7054715 DOI: 10.1016/j.ymthe.2019.11.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are widely used in clinical gene therapy to correct genetic disease by in vivo gene transfer. Although the vectors are useful, in part because of their limited immunogenicity, immune responses directed at vector components have complicated applications in humans. These include, for instance, innate immune sensing of vector components by plasmacytoid dendritic cells (pDCs), which sense the vector DNA genome via Toll-like receptor 9. Adaptive immune responses employ antigen presentation by conventional dendritic cells (cDCs), which leads to cross-priming of capsid-specific CD8+ T cells. In this study, we sought to determine the mechanisms that promote licensing of cDCs, which is requisite for CD8+ T cell activation. Blockage of type 1 interferon (T1 IFN) signaling by monoclonal antibody therapy prevented cross-priming. Furthermore, experiments in cell-type-restricted knockout mice showed a specific requirement for the receptor for T1 IFN (IFNaR) in cDCs. In contrast, natural killer (NK) cells are not needed, indicating a direct rather than indirect effect of T1 IFN on cDCs. In addition, co-stimulation by CD4+ T cells via CD40-CD40L was required for cross-priming, and blockage of co-stimulation but not of T1 IFN additionally reduced antibody formation against capsid. These mechanistic insights inform the development of targeted immune interventions.
Collapse
Affiliation(s)
- Jamie L Shirley
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | | | | | - Irene Zolotukhin
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - David M Markusic
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brad E Hoffman
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - Laurence M Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Mark A Wallet
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Department Pediatrics, University of Florida, Gainesville, FL, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
37
|
Nyalwidhe JO, Jurczyk A, Satish B, Redick S, Qaisar N, Trombly MI, Vangala P, Racicot R, Bortell R, Harlan DM, Greiner DL, Brehm MA, Nadler JL, Wang JP. Proteomic and Transcriptional Profiles of Human Stem Cell-Derived β Cells Following Enteroviral Challenge. Microorganisms 2020; 8:microorganisms8020295. [PMID: 32093375 PMCID: PMC7074978 DOI: 10.3390/microorganisms8020295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 01/04/2023] Open
Abstract
Enteroviral infections are implicated in islet autoimmunity and type 1 diabetes (T1D) pathogenesis. Significant β-cell stress and damage occur with viral infection, leading to cells that are dysfunctional and vulnerable to destruction. Human stem cell-derived β (SC-β) cells are insulin-producing cell clusters that closely resemble native β cells. To better understand the events precipitated by enteroviral infection of β cells, we investigated transcriptional and proteomic changes in SC-β cells challenged with coxsackie B virus (CVB). We confirmed infection by demonstrating that viral protein colocalized with insulin-positive SC-β cells by immunostaining. Transcriptome analysis showed a decrease in insulin gene expression following infection, and combined transcriptional and proteomic analysis revealed activation of innate immune pathways, including type I interferon (IFN), IFN-stimulated genes, nuclear factor-kappa B (NF-κB) and downstream inflammatory cytokines, and major histocompatibility complex (MHC) class I. Finally, insulin release by CVB4-infected SC-β cells was impaired. These transcriptional, proteomic, and functional findings are in agreement with responses in primary human islets infected with CVB ex vivo. Human SC-β cells may serve as a surrogate for primary human islets in virus-induced diabetes models. Because human SC-β cells are more genetically tractable and accessible than primary islets, they may provide a preferred platform for investigating T1D pathogenesis and developing new treatments.
Collapse
Affiliation(s)
- Julius O. Nyalwidhe
- Department of Microbiology and Molecular Cell Biology and Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23501, USA; (J.O.N.); (J.L.N.)
| | - Agata Jurczyk
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (A.J.); (S.R.); (R.B.); (D.L.G.); (M.A.B.)
| | - Basanthi Satish
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
| | - Sambra Redick
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (A.J.); (S.R.); (R.B.); (D.L.G.); (M.A.B.)
| | - Natasha Qaisar
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
| | - Melanie I. Trombly
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
| | - Pranitha Vangala
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA;
| | - Riccardo Racicot
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
| | - Rita Bortell
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (A.J.); (S.R.); (R.B.); (D.L.G.); (M.A.B.)
| | - David M. Harlan
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
| | - Dale L. Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (A.J.); (S.R.); (R.B.); (D.L.G.); (M.A.B.)
| | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (A.J.); (S.R.); (R.B.); (D.L.G.); (M.A.B.)
| | - Jerry L. Nadler
- Department of Microbiology and Molecular Cell Biology and Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23501, USA; (J.O.N.); (J.L.N.)
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Jennifer P. Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; (B.S.); (N.Q.); (M.I.T.); (R.R.); (D.M.H.)
- Correspondence: ; Tel.: +01-508-856-8414
| |
Collapse
|
38
|
Flynn JK, Dankers W, Morand EF. Could GILZ Be the Answer to Glucocorticoid Toxicity in Lupus? Front Immunol 2019; 10:1684. [PMID: 31379872 PMCID: PMC6652235 DOI: 10.3389/fimmu.2019.01684] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GC) are used globally to treat autoimmune and inflammatory disorders. Their anti-inflammatory actions are mainly mediated via binding to the glucocorticoid receptor (GR), creating a GC/GR complex, which acts in both the cytoplasm and nucleus to regulate the transcription of a host of target genes. As a result, signaling pathways such as NF-κB and AP-1 are inhibited, and cell activation, differentiation and survival and cytokine and chemokine production are suppressed. However, the gene regulation by GC can also cause severe side effects in patients. Systemic lupus erythematosus (SLE or lupus) is a multisystem autoimmune disease, characterized by a poorly regulated immune response leading to chronic inflammation and dysfunction of multiple organs, for which GC is the major current therapy. Long-term GC use, however, can cause debilitating adverse consequences for patients including diabetes, cardiovascular disease and osteoporosis and contributes to irreversible organ damage. To date, there is no alternative treatment which can replicate the rapid effects of GC across multiple immune cell functions, effecting disease control during disease flares. Research efforts have focused on finding alternatives to GC, which display similar immunoregulatory actions, without the devastating adverse metabolic effects. One potential candidate is the glucocorticoid-induced leucine zipper (GILZ). GILZ is induced by low concentrations of GC and is shown to mimic the action of GC in several inflammatory processes, reducing immunity and inflammation in in vitro and in vivo studies. Additionally, GILZ has, similar to the GC-GR complex, the ability to bind to both NF-κB and AP-1 as well as DNA directly, to regulate immune cell function, while potentially lacking the GC-related side effects. Importantly, in SLE patients GILZ is under-expressed and correlates negatively with disease activity, suggesting an important regulatory role of GILZ in SLE. Here we provide an overview of the actions and use of GC in lupus, and discuss whether the regulatory mechanisms of GILZ could lead to the development of a novel therapeutic for lupus. Increased understanding of the mechanisms of action of GILZ, and its ability to regulate immune events leading to lupus disease activity has important clinical implications for the development of safer anti-inflammatory therapies.
Collapse
Affiliation(s)
- Jacqueline K Flynn
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Wendy Dankers
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - Eric F Morand
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
39
|
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease whose pathogenesis can be conceptualized by a model based on a central role for immune complexes (ICs) between antinuclear antibodies and nucleic acids. According to this model, ICs can promote pathogenesis by two main mechanisms: deposition in the tissue to incite local inflammation and interaction with cells of the innate immune system to stimulate the production of cytokines, most prominently type 1 interferon. The latter stimulation results from the uptake of DNA and RNA in the form of ICs into cells and subsequent signaling by internal nucleic acid sensors for DNA and RNA. These sensors are likely important for the response to intracellular infection, although they may also be triggered during cell stress or injury by DNA or RNA aberrantly present in the cytoplasm. For IC formation, a source of extracellular DNA and RNA is essential. The current model of SLE posits that cell death is the origin of the nucleic acids in the ICs and that impairment of clearance mechanisms increases the amount of nuclear material in the extracellular space. This model of SLE is important since it points to new approaches to therapy; agents targeting interferon or the interferon receptor are examples of therapeutic approaches derived from this model. Future studies will explore novel biomarkers to monitor the operation of these mechanisms and to elucidate other steps in pathogenesis that can be targeted for therapy.
Collapse
Affiliation(s)
- David S Pisetsky
- Department of Medicine and Immunology, Duke University Medical Center and Medical Research Service, VA Medical Center, Durham, NC, USA
| |
Collapse
|
40
|
Sanghera C, Wong LM, Panahi M, Sintou A, Hasham M, Sattler S. Cardiac phenotype in mouse models of systemic autoimmunity. Dis Model Mech 2019; 12:dmm036947. [PMID: 30858306 PMCID: PMC6451423 DOI: 10.1242/dmm.036947] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Patients suffering from systemic autoimmune diseases are at significant risk of cardiovascular complications. This can be due to systemically increased levels of inflammation leading to accelerated atherosclerosis, or due to direct damage to the tissues and cells of the heart. Cardiac complications include an increased risk of myocardial infarction, myocarditis and dilated cardiomyopathy, valve disease, endothelial dysfunction, excessive fibrosis, and bona fide autoimmune-mediated tissue damage by autoantibodies or auto-reactive cells. There is, however, still a considerable need to better understand how to diagnose and treat cardiac complications in autoimmune patients. A range of inducible and spontaneous mouse models of systemic autoimmune diseases is available for mechanistic and therapeutic studies. For this Review, we systematically collated information on the cardiac phenotype in the most common inducible, spontaneous and engineered mouse models of systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis. We also highlight selected lesser-known models of interest to provide researchers with a decision framework to choose the most suitable model for their study of heart involvement in systemic autoimmunity.
Collapse
Affiliation(s)
- Chandan Sanghera
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Lok Man Wong
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Mona Panahi
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Amalia Sintou
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Muneer Hasham
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
41
|
Furie R, Werth VP, Merola JF, Stevenson L, Reynolds TL, Naik H, Wang W, Christmann R, Gardet A, Pellerin A, Hamann S, Auluck P, Barbey C, Gulati P, Rabah D, Franchimont N. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus erythematosus. J Clin Invest 2019; 129:1359-1371. [PMID: 30645203 DOI: 10.1172/jci124466] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Plasmacytoid DCs (pDC) produce large amounts of type I IFN (IFN-I), cytokines convincingly linked to systemic lupus erythematosus (SLE) pathogenesis. BIIB059 is a humanized mAb that binds blood DC antigen 2 (BDCA2), a pDC-specific receptor that inhibits the production of IFN-I and other inflammatory mediators when ligated. A first-in-human study was conducted to assess safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) effects of single BIIB059 doses in healthy volunteers (HV) and patients with SLE with active cutaneous disease as well as proof of biological activity and preliminary clinical response in the SLE cohort. METHODS A randomized, double-blind, placebo-controlled clinical trial was conducted in HV (n = 54) and patients with SLE (n = 12). All subjects were monitored for adverse events. Serum BIIB059 concentrations, BDCA2 levels on pDCs, and IFN-responsive biomarkers in whole blood and skin biopsies were measured. Skin disease activity was determined using the Cutaneous Lupus Erythematosus Disease Area and Severity Index Activity (CLASI-A). RESULTS Single doses of BIIB059 were associated with favorable safety and PK profiles. BIIB059 administration led to BDCA2 internalization on pDCs, which correlated with circulating BIIB059 levels. BIIB059 administration in patients with SLE decreased expression of IFN response genes in blood, normalized MxA expression, reduced immune infiltrates in skin lesions, and decreased CLASI-A score. CONCLUSIONS Single doses of BIIB059 were associated with favorable safety and PK/PD profiles and robust target engagement and biological activity, supporting further development of BIIB059 in SLE. The data suggest that targeting pDCs may be beneficial for patients with SLE, especially those with cutaneous manifestations. TRIAL REGISTRATION ClinicalTrials.gov NCT02106897. FUNDING Biogen Inc.
Collapse
Affiliation(s)
- Richard Furie
- Division of Rheumatology, Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | - Victoria P Werth
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania and Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Joseph F Merola
- Department of Dermatology and Department of Medicine, Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Merrill JT, Furie R, Werth VP, Khamashta M, Drappa J, Wang L, Illei G, Tummala R. Anifrolumab effects on rash and arthritis: impact of the type I interferon gene signature in the phase IIb MUSE study in patients with systemic lupus erythematosus. Lupus Sci Med 2018; 5:e000284. [PMID: 30588322 PMCID: PMC6280909 DOI: 10.1136/lupus-2018-000284] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/13/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
Abstract
Objective This post hoc analysis compared anifrolumab 300 mg every 4 weeks with placebo on rash and arthritis measures with different stringency in patients with moderate to severe SLE (phase IIb; MUSE; NCT01438489). Subgroups were analysed by type I interferon gene signature (IFNGS test–high or test–low). Methods Rash was measured with the SLE Disease Activity Index 2000 (SLEDAI-2K), British Isles Lupus Assessment Group (BILAG) Index and modified Cutaneous Lupus Erythematosus Disease Area and Severity Index (mCLASI). Arthritis was evaluated using SLEDAI-2K, BILAG and swollen and tender joint counts. Outcomes were measured at week 52. Results More anifrolumab-treated patients demonstrated resolution of rash by SLEDAI-2K versus placebo: 39/88 (44.3%) versus 13/88 (14.8%), OR (90% CI) 4.56 (2.48 to 8.39), p<0.001; improvement of BILAG: 48/82 (58.5%) versus 24/85 (28.2%), OR (90% CI) 3.59 (2.08 to 6.19), p<0.001; and ≥50% improvement by mCLASI: 57/92 (62.0%) versus 30/89 (33.7%), OR (90% CI) 3.31 (1.97 to 5.55), p<0.001. More anifrolumab-treated patients had improved arthritis by SLEDAI-2K versus placebo: 55/97 (56.7%) versus 42/99 (42.4%), OR (90% CI) 1.88 (1.16 to 3.04), p=0.032; and BILAG: 65/94 (69.1%) versus 47/95 (49.5%), OR (90% CI) 2.47 (1.48 to 4.12), p=0.003; and mean (SD) swollen and tender joint reductions: –5.5 (6.3) versus –3.4 (5.9), p=0.004. Comparable results were demonstrated in IFNGS test–high patients (n=151). In IFNGS test–low patients (n=50), substantial numerical differences in partial rash and arthritis responses were observed in anifrolumab-treated patients versus placebo, with statistical significance only for rash by BILAG in this small population. Conclusions Anifrolumab treatment was associated with improvements versus placebo in specific SLE features of arthritis and rash using measures of different stringency. Although driven by robust data in the prevalent IFNGS test–high population, further evaluation in IFNGS test–low patients is warranted.
Collapse
Affiliation(s)
- Joan T Merrill
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Richard Furie
- Division of Rheumatology, Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA
| | - Munther Khamashta
- Rheumatology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Jorn Drappa
- Research and Development, MedImmune, LLC, Gaithersburg, Maryland, USA
| | - Liangwei Wang
- Biometrics & Information Sciences, AstraZeneca, Gaithersburg, Maryland, USA
| | - Gabor Illei
- Clinical Development, MedImmune, LLC, Gaithersburg, Maryland, USA
| | - Raj Tummala
- Inflammation, Autoimmunity & Neuroscience, Global Medicines Development, AstraZeneca, Gaithersburg, Maryland, USA
| |
Collapse
|
43
|
Merrill JT, Furie R, Werth VP, Khamashta M, Drappa J, Wang L, Illei G, Tummala R. Anifrolumab effects on rash and arthritis: impact of the type I interferon gene signature in the phase IIb MUSE study in patients with systemic lupus erythematosus. Lupus Sci Med 2018; 5:e000284. [PMID: 30588322 DOI: 10.1136/lupus-2019-lsm.98] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/13/2018] [Accepted: 10/02/2018] [Indexed: 05/26/2023]
Abstract
OBJECTIVE This post hoc analysis compared anifrolumab 300 mg every 4 weeks with placebo on rash and arthritis measures with different stringency in patients with moderate to severe SLE (phase IIb; MUSE; NCT01438489). Subgroups were analysed by type I interferon gene signature (IFNGS test-high or test-low). METHODS Rash was measured with the SLE Disease Activity Index 2000 (SLEDAI-2K), British Isles Lupus Assessment Group (BILAG) Index and modified Cutaneous Lupus Erythematosus Disease Area and Severity Index (mCLASI). Arthritis was evaluated using SLEDAI-2K, BILAG and swollen and tender joint counts. Outcomes were measured at week 52. RESULTS More anifrolumab-treated patients demonstrated resolution of rash by SLEDAI-2K versus placebo: 39/88 (44.3%) versus 13/88 (14.8%), OR (90% CI) 4.56 (2.48 to 8.39), p<0.001; improvement of BILAG: 48/82 (58.5%) versus 24/85 (28.2%), OR (90% CI) 3.59 (2.08 to 6.19), p<0.001; and ≥50% improvement by mCLASI: 57/92 (62.0%) versus 30/89 (33.7%), OR (90% CI) 3.31 (1.97 to 5.55), p<0.001. More anifrolumab-treated patients had improved arthritis by SLEDAI-2K versus placebo: 55/97 (56.7%) versus 42/99 (42.4%), OR (90% CI) 1.88 (1.16 to 3.04), p=0.032; and BILAG: 65/94 (69.1%) versus 47/95 (49.5%), OR (90% CI) 2.47 (1.48 to 4.12), p=0.003; and mean (SD) swollen and tender joint reductions: -5.5 (6.3) versus -3.4 (5.9), p=0.004. Comparable results were demonstrated in IFNGS test-high patients (n=151). In IFNGS test-low patients (n=50), substantial numerical differences in partial rash and arthritis responses were observed in anifrolumab-treated patients versus placebo, with statistical significance only for rash by BILAG in this small population. CONCLUSIONS Anifrolumab treatment was associated with improvements versus placebo in specific SLE features of arthritis and rash using measures of different stringency. Although driven by robust data in the prevalent IFNGS test-high population, further evaluation in IFNGS test-low patients is warranted.
Collapse
Affiliation(s)
- Joan T Merrill
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Richard Furie
- Division of Rheumatology, Department of Medicine, Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA
| | - Munther Khamashta
- Rheumatology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Jorn Drappa
- Research and Development, MedImmune, LLC, Gaithersburg, Maryland, USA
| | - Liangwei Wang
- Biometrics & Information Sciences, AstraZeneca, Gaithersburg, Maryland, USA
| | - Gabor Illei
- Clinical Development, MedImmune, LLC, Gaithersburg, Maryland, USA
| | - Raj Tummala
- Inflammation, Autoimmunity & Neuroscience, Global Medicines Development, AstraZeneca, Gaithersburg, Maryland, USA
| |
Collapse
|
44
|
Mok CC. The Jakinibs in systemic lupus erythematosus: progress and prospects. Expert Opin Investig Drugs 2018; 28:85-92. [PMID: 30462559 DOI: 10.1080/13543784.2019.1551358] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, SAR China
| |
Collapse
|
45
|
Casey KA, Guo X, Smith MA, Wang S, Sinibaldi D, Sanjuan MA, Wang L, Illei GG, White WI. Type I interferon receptor blockade with anifrolumab corrects innate and adaptive immune perturbations of SLE. Lupus Sci Med 2018; 5:e000286. [PMID: 30538817 PMCID: PMC6257383 DOI: 10.1136/lupus-2018-000286] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/14/2018] [Accepted: 09/28/2018] [Indexed: 11/04/2022]
Abstract
Objective Anifrolumab is a fully human immunoglobulin G1 κ monoclonal antibody specific for subunit 1 of the type I interferon (IFN) α receptor. In a phase IIb study of adults with moderate to severe SLE, anifrolumab treatment demonstrated substantial reductions in multiple clinical endpoints. Here, we evaluated serum proteins and immune cells associated with SLE pathogenesis, type I interferon gene signature (IFNGS) test status and disease activity, and how anifrolumab affected these components. Methods Whole blood samples were collected from patients enrolled in MUSE (NCT01438489) for serum protein and cellular assessments at baseline and subsequent time points. Data were parsed by IFNGS test status (high/low) and disease activity. Protein expression and immune cell subsets were measured using multiplex immunoassay and flow cytometry, respectively. Blood samples from healthy donors were analysed for comparison. Results Baseline protein expression differed between patients with SLE and healthy donors, IFNGS test-high and -low patients, and patients with moderate and severe disease. Anifrolumab treatment lowered concentrations of IFN-induced chemokines associated with B, T and other immune cell migration in addition to proteins associated with endothelial activation that were dysregulated at baseline. IFNGS test-high patients and those with high disease activity were characterised by low baseline numbers of lymphocytes, circulating memory T-cell subsets and neutrophils. Anifrolumab treatment reversed lymphopenia and neutropenia in the total population, and normalised multiple T-cell subset counts in IFNGS test-high patients compared with placebo. Conclusions Anifrolumab treatment reversed IFN-associated changes at the protein and cellular level, indicating multiple modes of activity. Trial registration number NCT01438489.
Collapse
Affiliation(s)
- Kerry A Casey
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Xiang Guo
- Translational Sciences, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Michael A Smith
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Shiliang Wang
- Translational Sciences, MedImmune LLC, Gaithersburg, Maryland, USA
| | | | - Miguel A Sanjuan
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Liangwei Wang
- Biostatistics, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Gabor G Illei
- Clinical Development, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Wendy I White
- Research Bioinformatics, MedImmune LLC, Gaithersburg, Maryland, USA
| |
Collapse
|
46
|
Comprehensive assessment of the association between genes on JAK-STAT pathway (IFIH1, TYK2, IL-10) and systemic lupus erythematosus: a meta-analysis. Arch Dermatol Res 2018; 310:711-728. [DOI: 10.1007/s00403-018-1858-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/19/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
|
47
|
Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, Kinehara Y, Hirano T, Shima Y, Narazaki M, Kumanogoh A. Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis 2018; 77:1507-1515. [PMID: 29945921 PMCID: PMC6161667 DOI: 10.1136/annrheumdis-2018-212988] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/18/2018] [Accepted: 06/01/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Despite the importance of type I interferon (IFN-I) in systemic lupus erythematosus (SLE) pathogenesis, the mechanisms of IFN-I production have not been fully elucidated. Recognition of nucleic acids by DNA sensors induces IFN-I and interferon-stimulated genes (ISGs), but the involvement of cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) and stimulator of interferon genes (STING) in SLE remains unclear. We studied the role of the cGAS-STING pathway in the IFN-I-producing cascade driven by SLE serum. METHODS We collected sera from patients with SLE (n=64), patients with other autoimmune diseases (n=31) and healthy controls (n=35), and assayed them using a cell-based reporter system that enables highly sensitive detection of IFN-I and ISG-inducing activity. We used Toll-like receptor-specific reporter cells and reporter cells harbouring knockouts of cGAS, STING and IFNAR2 to evaluate signalling pathway-dependent ISG induction. RESULTS IFN-I bioactivity and ISG-inducing activities of serum were higher in patients with SLE than in patients with other autoimmune diseases or healthy controls. ISG-inducing activity of SLE sera was significantly reduced in STING-knockout reporter cells, and STING-dependent ISG-inducing activity correlated with disease activity. Double-stranded DNA levels were elevated in SLE. Apoptosis-derived membrane vesicles (AdMVs) from SLE sera had high ISG-inducing activity, which was diminished in cGAS-knockout or STING-knockout reporter cells. CONCLUSIONS AdMVs in SLE serum induce IFN-I production through activation of the cGAS-STING pathway. Thus, blockade of the cGAS-STING axis represents a promising therapeutic target for SLE. Moreover, our cell-based reporter system may be useful for stratifying patients with SLE with high ISG-inducing activity.
Collapse
Affiliation(s)
- Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - JeongHoon Park
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan.,Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan
| | - Hachirou Konaka
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - Wataru Aoki
- Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan.,Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Syunsuke Aburaya
- Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan.,Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Mitsuyoshi Ueda
- Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan.,Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan.,Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan
| | - Yoshitomo Hayama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - Yuhei Kinehara
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
| | - Toru Hirano
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan.,Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan.,Japan Science and Technology-Core Research for Evolutional Science and Technology (JST-CREST), Osaka University, Osaka, Japan
| |
Collapse
|
48
|
Riggs JM, Hanna RN, Rajan B, Zerrouki K, Karnell JL, Sagar D, Vainshtein I, Farmer E, Rosenthal K, Morehouse C, de Los Reyes M, Schifferli K, Liang M, Sanjuan MA, Sims GP, Kolbeck R. Characterisation of anifrolumab, a fully human anti-interferon receptor antagonist antibody for the treatment of systemic lupus erythematosus. Lupus Sci Med 2018; 5:e000261. [PMID: 29644082 PMCID: PMC5890856 DOI: 10.1136/lupus-2018-000261] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/02/2018] [Accepted: 03/08/2018] [Indexed: 01/19/2023]
Abstract
Objective We investigated the mechanistic and pharmacological properties of anifrolumab, a fully human, effector-null, anti-type I interferon (IFN) alpha receptor 1 (IFNAR1) monoclonal antibody in development for SLE. Methods IFNAR1 surface expression and internalisation on human monocytes before and after exposure to anifrolumab were assessed using confocal microscopy and flow cytometry. The effects of anifrolumab on type I IFN pathway activation were assessed using signal transducer and activator of transcription 1 (STAT1) phosphorylation, IFN-stimulated response element-luciferase reporter cell assays and type I IFN gene signature induction. The ability of anifrolumab to inhibit plasmacytoid dendritic cell (pDC) function and plasma cell differentiation was assessed by flow cytometry and ELISA. Effector-null properties of anifrolumab were assessed in antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays with B cells. Results Anifrolumab reduced cell surface IFNAR1 by eliciting IFNAR1 internalisation. Anifrolumab blocked type I IFN-dependent STAT1 phosphorylation and IFN-dependent signalling induced by recombinant and pDC-derived type I IFNs and serum of patients with SLE. Anifrolumab suppressed type I IFN production by blocking the type I IFN autoamplification loop and inhibited proinflammatory cytokine induction and the upregulation of costimulatory molecules on stimulated pDCs. Blockade of IFNAR1 suppressed plasma cell differentiation in pDC/B cell co-cultures. Anifrolumab did not exhibit CDC or ADCC activity. Conclusions Anifrolumab potently inhibits type I IFN-dependent signalling, including the type I IFN autoamplification loop, and is a promising therapeutic for patients with SLE and other diseases that exhibit chronic dysfunctional type I IFN signalling.
Collapse
Affiliation(s)
- Jeffrey M Riggs
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Richard N Hanna
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Bhargavi Rajan
- Clinical Pharmacology and DMPK, MedImmune LLC, Mountain View, California, USA
| | - Kamelia Zerrouki
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Jodi L Karnell
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Divya Sagar
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Inna Vainshtein
- Clinical Pharmacology and DMPK, MedImmune LLC, Mountain View, California, USA
| | - Erika Farmer
- Analytical Sciences, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Kimberly Rosenthal
- Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Chris Morehouse
- Translational Medicine, MedImmune LLC, Gaithersburg, Maryland, USA
| | | | - Kevin Schifferli
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Meina Liang
- Clinical Pharmacology and DMPK, MedImmune LLC, Mountain View, California, USA
| | - Miguel A Sanjuan
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Gary P Sims
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| | - Roland Kolbeck
- Respiratory, Inflammation and Autoimmunity, MedImmune LLC, Gaithersburg, Maryland, USA
| |
Collapse
|
49
|
Type I interferons promote the survival and proinflammatory properties of transitional B cells in systemic lupus erythematosus patients. Cell Mol Immunol 2018; 16:367-379. [PMID: 29563616 DOI: 10.1038/s41423-018-0010-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 01/02/2023] Open
Abstract
A hallmark of systemic lupus erythematosus (SLE) is the breaking of B-cell tolerance with the generation of high-affinity autoantibodies; however, the antibody-independent features of the B-cell compartment in SLE are less understood. In this study, we performed an extensive examination of B-cell subsets and their proinflammatory properties in a Chinese cohort of new-onset SLE patients. We observed that SLE patients exhibited an increased frequency of transitional B cells compared with healthy donors and rheumatoid arthritis patients. Plasma from SLE patients potently promoted the survival of transitional B cells in a type I IFN-dependent manner, which can be recapitulated by direct IFN-α treatment. Furthermore, the effect of IFN-α on enhanced survival of transitional B cells was associated with NF-κB pathway activation and reduced expression of the pro-apoptotic molecule Bax. Transitional B cells from SLE patients harbored a higher capacity to produce proinflammatory cytokine IL-6, which was also linked to the overactivated type I IFN pathway. In addition, the frequency of IL-6-producing transitional B cells was positively correlated with disease activity in SLE patients, and these cells were significantly reduced after short-term standard therapies. Thus, the current study provides a direct link between type I IFN pathway overactivation and the abnormally high frequency and proinflammatory properties of transitional B cells in active SLE patients, which contributes to the understanding of the roles of type I IFNs and B cells in the pathogenesis of SLE.
Collapse
|
50
|
Furie R, Wang L, Illei G, Drappa J. Systemic Lupus Erythematosus (SLE) Responder Index response is associated with global benefit for patients with SLE. Lupus 2018; 27:955-962. [DOI: 10.1177/0961203318758506] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- R Furie
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, NY, USA
| | - L Wang
- MedImmune, LLC, Gaithersburg, MD, USA
| | - G Illei
- MedImmune, LLC, Gaithersburg, MD, USA
| | - J Drappa
- MedImmune, LLC, Gaithersburg, MD, USA
| |
Collapse
|