1
|
Oftedal BE, Sjøgren T, Wolff ASB. Interferon autoantibodies as signals of a sick thymus. Front Immunol 2024; 15:1327784. [PMID: 38455040 PMCID: PMC10917889 DOI: 10.3389/fimmu.2024.1327784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.
Collapse
Affiliation(s)
- Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S. B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
2
|
Abdelhamid L, Mao J, Cabana-Puig X, Zhu J, Swartwout BK, Edwards MR, Testerman JC, Michaelis JS, Allen IC, Ahmed SA, Luo XM. Nlrp12 deficiency alters gut microbiota and ameliorates Faslpr-mediated systemic autoimmunity in male mice. Front Immunol 2023; 14:1120958. [PMID: 36969209 PMCID: PMC10036793 DOI: 10.3389/fimmu.2023.1120958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
NLRP12 has dual roles in shaping inflammation. We hypothesized that NLRP12 would modulate myeloid cells and T cell function to control systemic autoimmunity. Contrary to our hypothesis, the deficiency of Nlrp12 in autoimmune-prone B6.Faslpr/lpr mice ameliorated autoimmunity in males but not females. Nlrp12 deficiency dampened B cell terminal differentiation, germinal center reaction, and survival of autoreactive B cells leading to decreased production of autoantibodies and reduced renal deposition of IgG and complement C3. In parallel, Nlrp12 deficiency reduced the expansion of potentially pathogenic T cells, including double-negative T cells and T follicular helper cells. Furthermore, reduced pro-inflammatory innate immunity was observed, where the gene deletion decreased in-vivo expansion of splenic macrophages and mitigated ex-vivo responses of bone marrow-derived macrophages and dendritic cells to LPS stimulation. Interestingly, Nlrp12 deficiency altered the diversity and composition of fecal microbiota in both male and female B6/lpr mice. Notably, however, Nlrp12 deficiency significantly modulated small intestinal microbiota only in male mice, suggesting that the sex differences in disease phenotype might be gut microbiota-dependent. Together, these results suggest a potential pathogenic role of NLRP12 in promoting systemic autoimmunity in males. Future studies will investigate sex-based mechanisms through which NLRP12 differentially modulates autoimmune outcomes.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Department of Microbiology, College of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Jiangdi Mao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jing Zhu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Brianna K. Swartwout
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michael R. Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - James C. Testerman
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jacquelyn S. Michaelis
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, United States
| | - Irving Coy Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- *Correspondence: S. Ansar Ahmed, ; Xin M. Luo,
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- *Correspondence: S. Ansar Ahmed, ; Xin M. Luo,
| |
Collapse
|
3
|
Hasimoto M. [Gene-microbiota interactions for the development of systemic autoimmune diseases]. Nihon Yakurigaku Zasshi 2023; 158:313-318. [PMID: 37394552 DOI: 10.1254/fpj.22096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Genetics and gut microbiota contribute to the development of autoimmune diseases. SKG mice, which harbor a point mutation in the ZAP70 gene, develop autoimmune arthritis in BALB/c background and systemic lupus erythematosus in C57BL/6 background. Defective TCR signaling by ZAP70 mutation alters thymic selection thresholds and allows the positive selection of otherwise negatively selected self-reactive T cells. On the other hand, defective TCR signaling attenuates the positive selection of certain microbiota-reactive T cells, which lead to impaired IgA synthesis at mucosal site and gut dysbiosis. Gut dysbiosis, in turn, promotes autoimmunity via driving Th17 differentiation. Thus, defective TCR signaling leads to autoimmunity by altering thymic selection thresholds of self-reactive T cells and microbiota-reactive T cells. In this review, genomics-microbiota interactions for the development of autoimmunity will be discussed with the special focus on the recent finding obtained from animal models of autoimmunity with defective TCR signaling.
Collapse
Affiliation(s)
- Motomu Hasimoto
- Department of Clinical Immunology, Graduate School of Medicine, Osaka Metropolitan University
| |
Collapse
|
4
|
Becker YLC, Duvvuri B, Fortin PR, Lood C, Boilard E. The role of mitochondria in rheumatic diseases. Nat Rev Rheumatol 2022; 18:621-640. [PMID: 36175664 DOI: 10.1038/s41584-022-00834-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
The mitochondrion is an intracellular organelle thought to originate from endosymbiosis between an ancestral eukaryotic cell and an α-proteobacterium. Mitochondria are the powerhouses of the cell, and can control several important processes within the cell, such as cell death. Conversely, dysregulation of mitochondria possibly contributes to the pathophysiology of several autoimmune diseases. Defects in mitochondria can be caused by mutations in the mitochondrial genome or by chronic exposure to pro-inflammatory cytokines, including type I interferons. Following the release of intact mitochondria or mitochondrial components into the cytosol or the extracellular space, the bacteria-like molecular motifs of mitochondria can elicit pro-inflammatory responses by the innate immune system. Moreover, antibodies can target mitochondria in autoimmune diseases, suggesting an interplay between the adaptive immune system and mitochondria. In this Review, we discuss the roles of mitochondria in rheumatic diseases such as systemic lupus erythematosus, antiphospholipid syndrome and rheumatoid arthritis. An understanding of the different contributions of mitochondria to distinct rheumatic diseases or manifestations could permit the development of novel therapeutic strategies and the use of mitochondria-derived biomarkers to inform pathogenesis.
Collapse
Affiliation(s)
- Yann L C Becker
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Paul R Fortin
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Eric Boilard
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada.
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada.
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
5
|
Ma J, Gong Q, Pan X, Guo P, He L, You Y. Depletion of Fractalkine ameliorates renal injury and Treg cell apoptosis via the p38MAPK pathway in lupus-prone mice. Exp Cell Res 2021; 405:112704. [PMID: 34126056 DOI: 10.1016/j.yexcr.2021.112704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 11/20/2022]
Abstract
Fractalkine (FKN) is a chemokine with several roles, including chemotaxis; adhesion; and immune damage, which also participates in cell inflammation and apoptosis and responds to the pathogenesis of autoimmune diseases. Given the involvement of regulatory T cells (Treg) cells in autoimmune diseases, this study investigated the regulatory mechanism of FKN in renal injury and Treg apoptosis via the p38 mitogen-activated protein kinase (p38MAPK) signaling pathway in lupus-prone mice. Lupus was induced in BALB/c female mice by injection of pristane, followed by isolation of CD4+CD25+ Treg cells from the spleen of lupus model mice. To deplete FKN, mice received injection of an anti-FKN antibody, and Treg cells were transfected with FKN small-interfering RNA. Lupus mice and Treg cells were treated with the p38MAPK inhibitor SB203580 and activator U-46619, respectively, and urine protein and serum urea nitrogen, creatinine, and autoantibodies were measured and renal histopathological changes analyzed. We determined levels of FKN, phosphorylated p38 (p-p38), and forkhead box P3 (FOXP3) in renal tissue and Treg cells, and analyzed apoptosis rates and levels of key apoptotic factors in Treg cells. The renal FKN and p-p38 levels increased, whereas renal FOXP3 level decreased in lupus-prone mice. Treatment with the anti-FKN antibody and the p38MAPK inhibitor ameliorated proteinuria and renal function, significantly reducing serum autoantibody, renal FKN, and p-p38 levels while increasing renal FOXP3 level in lupus-prone mice. Moreover, FKN knockdown and administration of the p38MAPK inhibitor reduced apoptosis and levels of pro-apoptotic factors, increased levels of anti-apoptotic factors, and suppressed activation of p38MAPK signaling in Treg cells derived from lupus model mice. Furthermore, treatment with the p38MAPK activator U-46619 had the opposite effect on these cells. These data indicated that depletion of FKN ameliorated renal injury and Treg cell apoptosis via inhibition of p38MAPK signaling in lupus nephritis, suggesting that targeting FKN represents a potential therapeutic strategy for treating Lupus nephritis.
Collapse
Affiliation(s)
- Jingxue Ma
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Qiming Gong
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Xiuhong Pan
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Pengwei Guo
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Linlin He
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China
| | - Yanwu You
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
6
|
Yang SK, Zhang HR, Shi SP, Zhu YQ, Song N, Dai Q, Zhang W, Gui M, Zhang H. The Role of Mitochondria in Systemic Lupus Erythematosus: A Glimpse of Various Pathogenetic Mechanisms. Curr Med Chem 2020; 27:3346-3361. [PMID: 30479205 DOI: 10.2174/0929867326666181126165139] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is a polysystem autoimmune disease that adversely affects human health. Various organs can be affected, including the kidney or brain. Traditional treatment methods for SLE primarily rely on glucocorticoids and immunosuppressors. Unfortunately, these therapeutic agents cannot prevent a high recurrence rate after SLE remission. Therefore, novel therapeutic targets are urgently required. METHODS A systematic search of the published literature regarding the abnormal structure and function of mitochondria in SLE and therapies targeting mitochondria was performed in several databases. RESULTS Accumulating evidence indicates that mitochondrial dysfunction plays important roles in the pathogenesis of SLE, including influencing mitochondrial DNA damage, mitochondrial dynamics change, abnormal mitochondrial biogenesis and energy metabolism, mitophagy, oxidative stress, inflammatory reactions, apoptosis and NETosis. Further investigation of mitochondrial pathophysiological roles will result in further clarification of SLE. Specific lupus-induced organ damage also exhibits characteristic mitochondrial changes. CONCLUSION This review aimed to summarize the current research on the role of mitochondrial dysfunction in SLE, which will necessarily provide potential novel therapeutic targets for SLE.
Collapse
Affiliation(s)
- Shi-Kun Yang
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao-Ran Zhang
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Shu-Peng Shi
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Ying-Qiu Zhu
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Na Song
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qing Dai
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Gui
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Nephrology and rheumatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
TMEM203 is a binding partner and regulator of STING-mediated inflammatory signaling in macrophages. Proc Natl Acad Sci U S A 2019; 116:16479-16488. [PMID: 31346090 PMCID: PMC6697806 DOI: 10.1073/pnas.1901090116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Activators of interferons have received a great deal of interest in recent decades, both due to the central role they play in host defense against a range of pathogens, as well as the now well-recognized importance of dysregulated interferon activation/signaling in the pathogenesis of a number of highly prevalent and hard-to-treat diseases, such as systemic lupus erythematosus (SLE). Therefore, novel regulators of interferon activation are being sought as they may provide better targets to treat these diseases. We report the discovery of TMEM203 as an SLE-associated gene and a regulator of ligand-dependent activation of interferon production via STING. Thus, our work could form the basis of a novel therapeutic strategy for the treatment of interferonopathies, including SLE. Regulation of IFN signaling is critical in host recognition and response to pathogens while its dysregulation underlies the pathogenesis of several chronic diseases. STimulator of IFN Genes (STING) has been identified as a critical mediator of IFN inducing innate immune pathways, but little is known about direct coregulators of this protein. We report here that TMEM203, a conserved putative transmembrane protein, is an intracellular regulator of STING-mediated signaling. We show that TMEM203 interacts, functionally cooperates, and comigrates with STING following cell stimulation, which in turn leads to the activation of the kinase TBK1, and the IRF3 transcription factor. This induces target genes in macrophages, including IFN-β. Using Tmem203 knockout bone marrow-derived macrophages and transient knockdown of TMEM203 in human monocyte-derived macrophages, we show that TMEM203 protein is required for cGAMP-induced STING activation. Unlike STING, TMEM203 mRNA levels are elevated in T cells from patients with systemic lupus erythematosus, a disease characterized by the overexpression of type I interferons. Moreover, TMEM203 mRNA levels are associated with disease activity, as assessed by serum levels of the complement protein C3. Identification of TMEM203 sheds light into the control of STING-mediated innate immune responses, providing a potential novel mechanism for therapeutic interventions in STING-associated inflammatory diseases.
Collapse
|
8
|
Matsuo T, Hashimoto M, Sakaguchi S, Sakaguchi N, Ito Y, Hikida M, Tsuruyama T, Sakai K, Yokoi H, Shirakashi M, Tanaka M, Ito H, Yoshifuji H, Ohmura K, Fujii T, Mimori T. Strain-Specific Manifestation of Lupus-like Systemic Autoimmunity Caused byZap70Mutation. THE JOURNAL OF IMMUNOLOGY 2019; 202:3161-3172. [DOI: 10.4049/jimmunol.1801159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
|
9
|
Zhu Z, Yang C, Wen L, Liu L, Zuo X, Zhou F, Gao J, Zheng X, Shi Y, Zhu C, Liang B, Yin X, Wang W, Cheng H, Shen S, Tang X, Tang H, Sun L, Zhang A, Yang S, Cui Y, Zhang X, Sheng Y. RETRACTED: Bach2 regulates aberrant activation of B cell in systemic lupus erythematosus and can be negatively regulated by BCR-ABL/PI3K. Exp Cell Res 2018; 365:138-144. [PMID: 29501569 DOI: 10.1016/j.yexcr.2018.02.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/20/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). The article has been retracted at the request of the Editor-in-Chief and the authors. The journal is retracting this paper after the authors reached out to the journal with a statement that several images contained duplications from another published paper. Part of the panels in Figure 1D and Figure 4D are duplications of panels in Figure 7 of Wang et al., Int J Mol Sci (2016), DOI: 10.3390/ijms17060969. The β-actin panel in Figure 2E is same as the α-tubulin panel in Figure 4B. In addition, the corresponding author informed the journal that “there were serious conflicts of personal interest, part of co-authors of this paper were not involved in the study and not aware of the submission, and they did not authorize their names to appear in the article”.
Collapse
Affiliation(s)
- Zhengwei Zhu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Chao Yang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Leilei Wen
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Lu Liu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Xianbo Zuo
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Fusheng Zhou
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Jinping Gao
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Xiaodong Zheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Yinjuan Shi
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Caihong Zhu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Bo Liang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Xianyong Yin
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Wenjun Wang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Hui Cheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Songke Shen
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Xianfa Tang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Huayang Tang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Liangdan Sun
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Anping Zhang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Sen Yang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, East Street Cherry Park, Chaoyang District, Beijing 100029, China
| | - Xuejun Zhang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China
| | - Yujun Sheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230032, China.
| |
Collapse
|
10
|
Hong KM, Kim HK, Park SY, Poojan S, Kim MK, Sung J, Tsao BP, Grossman JM, Rullo OJ, Woo JMP, McCurdy DK, Rider LG, Miller FW, Song YW. CD3Z hypermethylation is associated with severe clinical manifestations in systemic lupus erythematosus and reduces CD3ζ-chain expression in T cells. Rheumatology (Oxford) 2017; 56:467-476. [PMID: 27940592 DOI: 10.1093/rheumatology/kew405] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Indexed: 11/14/2022] Open
Abstract
Objective The importance of hypomethylation in SLE is well recognized; however, the significance of hypermethylation has not been well characterized. We screened hypermethylated marks in SLE and investigated their possible implications. Methods DNA methylation marks were screened in SLE whole-blood DNA by microarray, and two marks ( CD3Z and VHL hypermethylations) were confirmed by a methylation single-base extension method in two independent ethnic cohorts consisting of 207 SLE patients and 151 controls. The correlation with clinical manifestations and the genetic influence on those epigenetic marks were analysed. Results Two epigenetic marks, CD3Z and VHL hypermethylation, were significantly correlated with SLE: CD3Z hypermethylation (odds ratio = 7.76; P = 1.71 × 10 -13 ) and VHL hypermethylation (odds ratio = 3.77; P = 3.20 × 10 -8 ), and the increased CD3Z methylation was correlated with downregulation of the CD3ζ-chain in SLE T cells. In addition, less genetic influence on CD3Z methylation relative to VHL methylation was found in analyses of longitudinal and twin samples. Furthermore, a higher CD3Z methylation level was significantly correlated with a higher SLE disease activity index and more severe clinical manifestations, such as proteinuria, haemolytic anaemia and thrombocytopenia, whereas VHL hypermethylation was not. Conclusion CD3Z hypermethylation is an SLE risk factor that can be modified by environmental factors and is associated with more severe SLE clinical manifestations, which are related to deranged T cell function by downregulating the CD3ζ-chain.
Collapse
Affiliation(s)
| | | | | | - Shiv Poojan
- Research Institute, National Cancer Center, Goyang
| | - Mi-Kyung Kim
- Research Institute, National Cancer Center, Goyang
| | - Joohon Sung
- Department of Epidemiology, School of Public Health, Seoul National University, Seoul, Korea
| | | | | | - Ornella J Rullo
- Division of Pediatric Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Jennifer M P Woo
- Division of Pediatric Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Deborah K McCurdy
- Division of Pediatric Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Lisa G Rider
- Department of Health and Human Services, Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Frederick W Miller
- Department of Health and Human Services, Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Yeong-Wook Song
- Department of Internal Medicine.,Department of Molecular Medicine and Biopharmaceutical Sciences, Medical Research Center, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
11
|
Yin J, Zhang J, Lu Q. The role of basic leucine zipper transcription factor E4BP4 in the immune system and immune-mediated diseases. Clin Immunol 2017; 180:5-10. [PMID: 28365317 DOI: 10.1016/j.clim.2017.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 01/03/2023]
Abstract
Basic leucine zipper transcription factor E4BP4 (also known as NFIL3) has been implicated in the molecular and cellular mechanisms of functions and activities in mammals. The interactions between E4BP4 and major regulators of cellular processes have triggered significant interest in the roles of E4BP4 in the pathogenesis of certain chronic diseases. Indeed, novel discoveries have been emerging to illustrate the involvement of E4BP4 in multiple disorders. It is recognized that E4BP4 is extensively involved in some immune-mediated diseases, but the mechanisms of E4BP4 involvement in these complex diseases remain poorly defined. Here we review the regulatory mechanisms of E4BP4 engaging in not only the biological function but also the development of immune-mediated diseases, paving the way for future therapies.
Collapse
Affiliation(s)
- Jinghua Yin
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139, Renmin Road, Changsha 410011, China
| | - Jian Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH43210, USA.
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, No. 139, Renmin Road, Changsha 410011, China.
| |
Collapse
|
12
|
Abstract
In a single assay, gene microarrays generate tens of thousands of measurements for the relative levels of messenger RNA expression, and thus hold promise to uncover the regulation of transcriptional responses behind clinical phenotypes of various diseases. Systemic lupus erythematosus (SLE) offers a unique opportunity to study gene expression both systemically and organ specific, as the tissues involved and specifically peripheral blood cells are readily accessible for molecular analysis. In the current review we highlight the current knowledge related to gene microarray in SLE. We approached the following questions: 1) Can gene microarray technology be used to translate molecular profiles into meaningful and applicable clinical information? 2) Does the assessment of differential gene expression provide specific signatures that may contribute to diagnostic and prognostic markers of SLE? 3) Can clinicians be helped in monitoring disease activity by identification of drug response gene profile? 4) Does evaluation of differential gene expression provide clues to detect previously unrecognized genes associated with the disease? It is evident that though not all questions can be currently answered appropriately, gene expression studies in SLE have important implications and will not only be beneficial for SLE patients, but will also lead to a better understanding of other autoimmune inflammatory diseases, thereby leading to novel diagnostic and therapeutic strategies in autoimmunity.
Collapse
Affiliation(s)
- M Mandel
- Blood Center, Sheba Medical Center, Tel-Hashomer, Israel
| | | |
Collapse
|
13
|
Kayser C, Alberto FL, da Silva NP, Andrade LEC. Decreased number of T cells bearing TCR rearrangement excision circles (TREC) in active recent onset systemic lupus erythematosus. Lupus 2016; 13:906-11. [PMID: 15645744 DOI: 10.1191/0961203304lu2031oa] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Systemic lupus erythematosus (SLE) is characterized by several T lymphocyte abnormalities. An indirect assessment of recent thymus emigrants (RTE) has been recently been made available by measuring the number of TCR recombination excision circles (TREC) in peripheral T cells. We studied TREC levels in peripheral blood mononuclear cells (PBMC) of 32 SLE patients with active disease and 32 normal age- and sex-matched controls. Signal-joint TREC concentration was determined by real-time quantitative-PCR as the number of TREC copies/μg PBMC DNA. SLE patients had lower TREC levels (4.1 ±3.9 ×104 TREC/μg DNA) than controls (8.9 ±7.9 ×104/μg DNA) ( P = 0.004). There was an inverse correlation between age and TREC levels in controls ( r = 20.41, P = 0.02) but not in SLE patients. No clinical association was observed between TREC levels and clinical and laboratory SLE manifestations. TREC levels tended to be lower in patients with SLEDAI above 20 than in the rest of the patients ( P = 0.08). The decreased PBMC TREC levels is indicative of a low proportion of RTE in SLE and could be caused by decreased RTE output and/or by increased peripheral T cell proliferation in this disease. The under-representation of RTE in the peripheral T cell pool may play a role in the immune tolerance abnormalities observed in SLE.
Collapse
Affiliation(s)
- C Kayser
- Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
14
|
Abstract
Preclinical studies have provided proof of concept for the feasibility and efficacy of gene therapy in human systemic lupus erythematosus (SLE). Successful efforts include gene constructs that alter the expression of cytokines or limit the cognate interaction of immune cells. Other efforts may include gene modified cell transfersuch as autologousB cells transfectedwith tolerogenicconstructsor T cells in which specific molecular aberrations have been corrected.
Collapse
Affiliation(s)
- V C Kyttaris
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | |
Collapse
|
15
|
Tang L, Wang Y, Zheng S, Bao M, Zhang Q, Li J. PTPN22 polymorphisms, but not R620W, were associated with the genetic susceptibility of systemic lupus erythematosus and rheumatoid arthritis in a Chinese Han population. Hum Immunol 2016; 77:692-698. [PMID: 27166176 DOI: 10.1016/j.humimm.2016.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/18/2016] [Accepted: 04/28/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The present study aimed to detect a possible association between PTPN22 gene polymorphisms and rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) in a Chinese Han population. METHODS 7 PTPN22 SNPs were genotyped in 358 patients with RA and 713 patients with SLE, as well as 564 RA controls and 672 SLE controls by Restriction Fragment Length Polymorphism (RFLP). Association analyses were conducted on the whole data set. Significant relationships were also examined between clinical features and SNPs for both RA and SLE. RESULTS Rs2476601 was lack of polymorphism with a ⩽0.1% frequency in both SLE and RA patients and healthy controls in our study. The two SNPs rs1217414 and rs3811021 of PTPN22 shown strong association with both SLE (rs1217414T: padj = 6.07e-004, OR=0.57; rs3811021C: padj = 4.68e-005, OR=0.65) and RA (rs1217414T: padj = 2.01e-008, OR=0.26; rs3811021C: padj = 0.028, OR=0.70). And the rs3765598 revealed a strong risk factor for SLE (p=9.38e-009, padj = 6.57e-008, OR=1.93), but not for RA (p=0.48, OR=1.12). Moreover, protective haplotype ACTTC in RA (p=7.73e-016, padj = 5.51-015, OR[95%CI]=0.02[0.002-0.10]) and SLE (p=8.29e-018, padj = 5.80e-017, OR[95%CI]=0.11[0.06-0.21]) were observed. In addition, the distribution of risk haplotypes ACGTC and GCTTT in RA (ACGTC: p=0.0006, padj = 0.004, OR[95%CI]=1.85[1.29-2.63]; GCTTT: p=2.62e-005, padj = 1.85e-004, OR[95%CI]=2.40[1.57-3.65]) and SLE (ACGTC: p=0.0006, padj = 0.004, OR[95%CI]=1.85[1.29-2.63]; ACGTC: p=7.74e-011, padj = 6.81e-010, OR[95%CI]=2.21[1.12-3.34]; GCTTT: p=2.40[1.57-3.65], padj = 2.26e-006, OR[95%CI]=2.64[1.79-3.87]) were significant different from that in controls. Furthermore, significant association was observed between the PTPN22 rs3765598 and antinuclear antibodies 1 (ANA1) in SLE. CONCLUSIONS Our data provide strong evidence that the rs1217414 and rs3811021 in PTPN22 gene might be common protective factors contributed to SLE and RA susceptibility in the Chinese Han population. While, the rs3765598 might increase the genetic susceptibility of SLE, but not RA.
Collapse
Affiliation(s)
- Liang Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, PR China; School of Basic Medical Science, Changsha Medical University, Changsha, PR China
| | - Yan Wang
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, PR China; School of Basic Medical Science, Changsha Medical University, Changsha, PR China
| | - Shui Zheng
- Key Laboratory for Fertility Regulation and Birth Health of Minority Nationalities of Yunnan Province, Judicial Expertise Center, Yunnan Population and Family Planning Research Institute, Kunming, PR China
| | - Meihua Bao
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, PR China; School of Basic Medical Science, Changsha Medical University, Changsha, PR China
| | - Qingsong Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, PR China; School of Basic Medical Science, Changsha Medical University, Changsha, PR China
| | - Jianming Li
- Xiangya Hospital, Central South University, Changsha, PR China; Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, PR China.
| |
Collapse
|
16
|
Song LJ, Wang X, Wang XP, Li D, Ding F, Liu HX, Yu X, Li XF, Shu Q. Increased Tim-3 expression on peripheral T lymphocyte subsets and association with higher disease activity in systemic lupus erythematosus. Diagn Pathol 2015; 10:71. [PMID: 26076826 PMCID: PMC4469310 DOI: 10.1186/s13000-015-0306-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 05/29/2015] [Indexed: 12/21/2022] Open
Abstract
Background Both the T cell immunoglobulin domain- and mucin domain-containing molecule-3 (Tim-3) and the death receptor Fas contribute to the pathogenesis of various autoimmune diseases, including systemic lupus erythematosus (SLE). The aim of the present study was to determine whether Tim-3 and Fas are co-expressed on certain peripheral T lymphocyte subsets, and whether this expression is associated with greater disease activity in SLE. Methods Peripheral blood mononuclear cells were isolated from 46 patients newly diagnosed with SLE and 28 age- and sex-matched healthy controls (HCs). Expression of Tim-3 and Fas on T subsets was analyzed by flow cytometry, while mRNA levels of the Tim-3 ligand galectin-9 and Fas ligand FasL were assayed using real-time RT-PCR. Results The proportions of CD3+CD4+ and CD3+CD4- T cells expressing Tim-3+ and Tim+Fas+ were significantly higher in patients than in HCs (p < 0.05), while the proportions of these subtypes expressing Fas were similar for the two groups. Patients with active SLE, as defined by their score on the SLE Disease Activity Index, had lower proportions of CD3+CD4+ T cells and higher proportions of CD3+CD4+Tim-3+ and CD3+CD4+Tim-3+Fas+ T cells than did patients with stable SLE. Serum levels of complement C3 and C4 proteins, considered as a marker of SLE activity, correlated negatively with proportions of CD3+CD4+ and CD3+CD4- T cells expressing Tim-3. Conclusions Expression of Tim-3 and co-expression of Tim-3 and Fas on certain peripheral T subsets are associated with disease activity in SLE patients. Future research should examine whether the same is true of other T subsets implicated in SLE, and should explore the potential role(s) of Tim-3 in the disease pathway. Virtual slides http://www.diagnosticpathology.diagnomx.eu/vs/1855527845145188
Collapse
Affiliation(s)
- Li-jun Song
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Xiao Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Xu-ping Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Ji'nan, 250012, China.
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Ji'nan, 250012, China.
| | - Feng Ding
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Hua-xiang Liu
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Xiao Yu
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Xing-fu Li
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| | - Qiang Shu
- Department of Rheumatology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Ji'nan, 250012, China.
| |
Collapse
|
17
|
Monsiváis-Urenda A, Gómez-Martin D, Santana-de-Anda K, Cruz-Martínez J, Alcocer-Varela J, González-Amaro R. Defective expression and function of the ILT2/CD85j regulatory receptor in dendritic cells from patients with systemic lupus erythematosus. Hum Immunol 2013; 74:1088-96. [DOI: 10.1016/j.humimm.2013.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 05/06/2013] [Accepted: 05/29/2013] [Indexed: 02/01/2023]
|
18
|
Differential effect of allogeneic versus syngeneic mesenchymal stem cell transplantation in MRL/lpr and (NZB/NZW)F1 mice. Clin Immunol 2012; 145:142-52. [DOI: 10.1016/j.clim.2012.08.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 11/18/2022]
|
19
|
Epstein-Barr virus and systemic lupus erythematosus. Clin Dev Immunol 2012; 2012:370516. [PMID: 22811739 PMCID: PMC3395176 DOI: 10.1155/2012/370516] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 03/23/2012] [Accepted: 04/14/2012] [Indexed: 11/18/2022]
Abstract
The etiology of SLE is not fully established. SLE is a disease with periods of waning disease activity and intermittent flares. This fits well in theory to a latent virus infection, which occasionally switches to lytic cycle, and EBV infection has for long been suspected to be involved. This paper reviews EBV immunobiology and how this is related to SLE pathogenesis by illustrating uncontrolled reactivation of EBV as a disease mechanism for SLE. Studies on EBV in SLE patients show enlarged viral load, abnormal expression of viral lytic genes, impaired EBV-specific T-cell response, and increased levels of EBV-directed antibodies. These results suggest a role for reactivation of EBV infection in SLE. The increased level of EBV antibodies especially comprises an elevated titre of IgA antibodies, and the total number of EBV-reacting antibody isotypes is also enlarged. As EBV is known to be controlled by cell-mediated immunity, the reduced EBV-specific T-cell response in SLE patients may result in defective control of EBV causing frequent reactivation and expression of lytic cycle antigens. This gives rise to enhanced apoptosis and amplified cellular waste load resulting in activation of an immune response and development of EBV-directed antibodies and autoantibodies to cellular antigens.
Collapse
|
20
|
|
21
|
Takeuchi T, Suzuki K, Kondo T, Yoshimoto K, Tsuzaka K. CD3 ζ defects in systemic lupus erythematosus. Ann Rheum Dis 2012; 71 Suppl 2:i78-81. [PMID: 22460144 DOI: 10.1136/annrheumdis-2011-200641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The prototype autoimmune disease, systemic lupus erythematosus (SLE), has been known to be associated with deficiency of ζ chain, a component of the T-cell receptor-CD3 complex. Comprehensive analysis has shown that expression of the CD3 ζ chain is attenuated or absent in over half of SLE patients. Furthermore, aberrant transcripts of the CD3 ζ chain, including spliced variants lacking exon 7 or having a short 3'-untranslated region, have been detected in SLE T cells. Although attenuated expression of the CD3 ζ chain is also observed in cancer patients, infections and other autoimmune diseases, sustained attenuation of the CD3 ζ expression accompanied with aberrant transcripts are only observed in SLE. In this study, the authors review the unique features of CD3 ζ defects observed in SLE and discuss the molecular basis of the defects by recent findings in animal models, single-nucleotide polymorphisms and genome-wide association studies.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Department of Internal Medicine, Division of Rheumatology, School of Medicine, Keio University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
22
|
Zhang Y, Huang W, Li F, Feng S, Kang K, Xu J. Octreotide for the treatment of systemic lupus erythematosus: clinical effects and an in vitro study on its therapeutic mechanism. Lupus 2012; 20:1172-81. [PMID: 21979989 DOI: 10.1177/0961203311409268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increased serum growth hormone (GH), together with high expression of growth hormone receptor on peripheral blood mononuclear cells (PBMCs), correlates with systemic lupus erythematosus (SLE) activity, suggesting that modulation of GH signaling may affect SLE activity. We explored the effects of octreotide (OCT), an analog of somatostatin that suppresses the release of GH, in SLE. The objectives of the study were to investigate effects of OCT on the proliferative capacity and cytokine expression of PBMCs from patients with SLE and to investigate therapeutic effects of OCT in patients with SLE. PBMCs from 13 active/inactive SLE patients and 11 controls were pretreated with or without GH and cultured with OCT. The proliferation of PBMCs was assessed by MTT assay and cytokines were quantified by ELISA. We compared the clinical response of 12 patients with SLE treated with OCT (100 µg twice daily) with 12 patients treated with prednisone over three months. OCT inhibited PBMC proliferation in a dose-dependent manner and decreased the secretion of interleukin-6 (IL-6), interleukin-10 (IL-10), and interferon-gamma (IFN-γ). Patients treated with OCT demonstrated improvements in SLEDAI, dsDNA titer, complement levels, and erythrocyte sedimentation rate (ESR). OCT inhibited PBMC proliferation and PBMC secretion of IL-6, IL-10 and IFN-γ stimulated by GH. Treatment of patients with OCT resulted in clinical improvement in SLE.
Collapse
Affiliation(s)
- Y Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
23
|
Pavón EJ, García-Rodríguez S, Zumaquero E, Perandrés-López R, Rosal-Vela A, Lario A, Longobardo V, Carrascal M, Abián J, Callejas-Rubio JL, Ortego-Centeno N, Zubiaur M, Sancho J. Increased expression and phosphorylation of the two S100A9 isoforms in mononuclear cells from patients with systemic lupus erythematosus: a proteomic signature for circulating low-density granulocytes. J Proteomics 2011; 75:1778-91. [PMID: 22230807 DOI: 10.1016/j.jprot.2011.12.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 11/03/2011] [Accepted: 12/14/2011] [Indexed: 10/14/2022]
Abstract
Proteins differentially expressed in peripheral blood mononuclear cells (PBMCs) from systemic lupus erythematosus (SLE) patients versus Normal controls were identified by 2-DE and MALDI-MS. Thus, S100A9 expression was significantly increased in SLE PBMCs relative to Normal PBMCs at both mRNA and protein levels. Increased S100A9 levels in SLE PBMCs correlated positively with the abnormal presence of low-density granulocytes (LDGs) detected by flow-cytometry in the mononuclear cell fractions. Another set of proteins that were differentially expressed in SLE PBMCs formed S100A9-independent clusters, suggesting that these differences in protein expression are in fact reflecting changes in the abundance of specific cell types. In SLE PBMCs spots of the two S100A9 isoforms, S100A9-l and S100A9-s, and their phosphorylated counterparts were identified and confirmed to be phosphorylated at Thr(113) by MS/MS analyses. In addition, the phorbol ester PMA alone or in combination with ionomycin induced a stronger increase in threonine phosphorylation of S100A9 in SLE than in Normal PBMCs, while the same stimuli caused the opposite effect on phosphorylation and activation of Erk1/2, suggesting the existence of an abnormal S100A9 signaling in SLE PBMCs. Therefore, the expansion and activation of LDGs in SLE seems to underlie this prominent S100A9 signature.
Collapse
Affiliation(s)
- Esther J Pavón
- Departamento de Biología Celular e Inmunología, Instituto de Parasitología y Biomedicina "López-Neyra", IPBLN-CSIC, Armilla, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hughes T, Sawalha AH. The role of epigenetic variation in the pathogenesis of systemic lupus erythematosus. Arthritis Res Ther 2011; 13:245. [PMID: 22044622 PMCID: PMC3308098 DOI: 10.1186/ar3484] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The focus of the present review is on the extent to which epigenetic alterations influence the development of systemic lupus erythematosus. Lupus is a systemic autoimmune disease characterized by the production of autoantibodies directed at nuclear self-antigens. A DNA methylation defect in CD4+ T cells has long been observed in idiopathic and drug-induced lupus. Recent studies utilizing high-throughput technologies have further characterized the nature of the DNA methylation defect in lupus CD4+ T cells. Emerging evidence in the literature is revealing an increasingly interconnected network of epigenetic dysregulation in lupus. Recent reports describe variable expression of a number of regulatory microRNAs in lupus CD4+ T cells, some of which govern the expression of DNA methyltransferase 1. While studies to date have revealed a significant role for epigenetic defects in the pathogenesis of lupus, the causal nature of epigenetic variation in lupus remains elusive. Future longitudinal epigenetic studies in lupus are therefore needed.
Collapse
Affiliation(s)
- Travis Hughes
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Amr H Sawalha
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- US Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- 825 NE 13th Street, MS#24, Oklahoma City, OK 73104, USA
| |
Collapse
|
25
|
Doníz-Padilla L, Martínez-Jiménez V, Niño-Moreno P, Abud-Mendoza C, Hernández-Castro B, González-Amaro R, Layseca-Espinosa E, Baranda-Cándido L. Expression and function of Cbl-b in T cells from patients with systemic lupus erythematosus, and detection of the 2126 A/G Cblb gene polymorphism in the Mexican mestizo population. Lupus 2011; 20:628-35. [PMID: 21558139 DOI: 10.1177/0961203310394896] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is characterized by abnormalities in the function of T and B lymphocytes and in the signaling pathways induced through their receptors. Cbl-b is an intracellular adaptor protein that plays a key role in the negative regulation of lymphocyte activity. We explored the expression and function of Cbl-b in T lymphocytes from SLE patients. In addition, the possible association of SLE and a single nucleotide polymorphism (SNP) of the Cblb gene was determined. We studied 150 SLE patients, 163 healthy individuals, and 14 patients with rheumatoid arthritis (RA). The expression of Cbl-b was analyzed in the peripheral blood mononuclear cells, and the negative regulatory function of Cbl-b was assessed by analyzing actin polymerization and the phosphorylation of JNK and c-Jun induced through CD3. Furthermore, the 2126(A/G) SNP of the Cblb gene was detected by real-time polymerase chain reaction. We found a significant small reduction in the expression of Cbl-b as well as increased levels of activation of c-Jun and actin polymerization in T lymphocytes from patients with SLE compared with healthy controls or RA patients. In addition, a significant association between the 2126(A/G) SNP and SLE was detected. Our data suggest that Cbl-b may contribute to the deregulated activation of T lymphocytes observed in SLE.
Collapse
Affiliation(s)
- L Doníz-Padilla
- Department of Immunology, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Sunahori K, Juang YT, Kyttaris VC, Tsokos GC. Promoter hypomethylation results in increased expression of protein phosphatase 2A in T cells from patients with systemic lupus erythematosus. THE JOURNAL OF IMMUNOLOGY 2011; 186:4508-17. [PMID: 21346232 DOI: 10.4049/jimmunol.1000340] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The catalytic subunit α isoform of protein phosphatase 2A (PP2Acα) activity, protein, and mRNA have been found increased in systemic lupus erythematosus (SLE) T cells and to contribute to decreased IL-2 production. The PP2Acα promoter activity is controlled epigenetically through the methylation of a CpG within a cAMP response element (CRE) motif defined by its promoter. We considered that hypomethylation may account for the increased expression of PP2Acα in patients with SLE. Using bisulfite sequencing, we found that SLE T cells displayed decreased DNA methylation in the promoter region compared with normal T cells. More importantly, we found that the CRE-defined CpG, which binds p-CREB, is significantly less methylated in SLE compared with normal T cells, and the levels of methylation correlated with decreased amounts of DNA methyltransferase 1 transcripts. Methylation intensity correlated inversely with levels of PP2Acα mRNA and SLE disease activity. Chromatin immunoprecipitation assays revealed more binding of p-CREB to the CRE site in SLE T cells, resulting in increased expression of PP2Acα. We propose that PP2Acα represents a new methylation-sensitive gene that, like the previously reported CD70 and CD11a, contributes to the pathogenesis of SLE.
Collapse
Affiliation(s)
- Katsue Sunahori
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
27
|
A New Epigenetic Challenge: Systemic Lupus Erythematosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 711:117-36. [DOI: 10.1007/978-1-4419-8216-2_9] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Juang YT, Peoples C, Kafri R, Kyttaris VC, Sunahori K, Kis-Toth K, Fitzgerald L, Ergin S, Finnell M, Tsokos GC. A systemic lupus erythematosus gene expression array in disease diagnosis and classification: a preliminary report. Lupus 2010; 20:243-9. [PMID: 21138984 DOI: 10.1177/0961203310383072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a clinically heterogeneous disease diagnosed on the presence of a constellation of clinical and laboratory findings. At the pathogenetic level, multiple factors using diverse biochemical and molecular pathways have been recognized. Succinct recognition and classification of clinical disease subsets, as well as the availability of disease biomarkers, remains largely unsolved. Based on information produced by the present authors' and other laboratories, a lupus gene expression array consisting of 30 genes, previously claimed to contribute to aberrant function of T cells, was developed. An additional eight genes were included as controls. Peripheral blood was obtained from 10 patients (19 samples) with SLE and six patients with rheumatoid arthritis (RA) as well as 19 healthy controls. T cell mRNA was subjected to reverse transcription and PCR, and the gene expression levels were measured. Conventional statistical analysis was performed along with principal component analysis (PCA) to capture the contribution of all genes to disease diagnosis and clinical parameters. The lupus gene expression array faithfully informed on the expression levels of genes. The recorded changes in expression reflect those reported in the literature by using a relatively small (5 ml) amount of peripheral blood. PCA of gene expression levels placed SLE samples apart from normal and RA samples regardless of disease activity. Individual principal components tended to define specific disease manifestations such as arthritis and proteinuria. Thus, a lupus gene expression array based on genes previously claimed to contribute to immune pathogenesis of SLE may define the disease, and principal components of the expression of 30 genes may define patients with specific disease manifestations.
Collapse
Affiliation(s)
- Y-T Juang
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Das UN. Current and emerging strategies for the treatment and management of systemic lupus erythematosus based on molecular signatures of acute and chronic inflammation. J Inflamm Res 2010; 3:143-70. [PMID: 22096364 PMCID: PMC3218729 DOI: 10.2147/jir.s9425] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lupus is a chronic, systemic inflammatory condition in which eicosanoids, cytokines, nitric oxide (NO), a deranged immune system, and genetics play a significant role. Our studies revealed that an imbalance in the pro- and antioxidants and NO and an alteration in the metabolism of essential fatty acids exist in lupus. The current strategy of management includes administration of nonsteroidal anti-inflammatory drugs such as hydroxychloroquine and immunosuppressive drugs such as corticosteroids. Investigational drugs include the following: 1) belimumab, a fully human monoclonal antibody that specifically recognizes and inhibits the biological activity of B-lymphocyte stimulator, also known as B-cell-activation factor of the TNF family; 2) stem cell transplantation; 3) rituximab, a chimeric monoclonal antibody against CD20, which is primarily found on the surface of B-cells and can therefore destroy B-cells; and 4) IL-27, which has potent anti-inflammatory actions. Our studies showed that a regimen of corticosteroids and cyclophosphamide, and methods designed to enhance endothelial NO synthesis and augment antioxidant defenses, led to induction of long-lasting remission of the disease. These results suggest that methods designed to modulate molecular signatures of the disease process and suppress inflammation could be of significant benefit in lupus. Some of these strategies could be vagal nerve stimulation, glucose-insulin infusion, and administration of lipoxins, resolvins, protectins, and nitrolipids by themselves or their stable synthetic analogs that are known to suppress inflammation and help in the resolution and healing of the inflammation-induced damage. These strategies are likely to be useful not only in lupus but also in other conditions, such as rheumatoid arthritis, scleroderma, ischemia-reperfusion injury to the myocardium, ischemic heart disease, and sepsis.
Collapse
Affiliation(s)
- Undurti N Das
- Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India; UND Life Sciences, Shaker Heights, OH, USA
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Epigenetic mechanisms regulate gene expression, and epigenetic gene dysregulation is implicated in the pathogenesis of a growing number of disorders. Of the autoimmune diseases, epigenetic mechanisms are most clearly involved in human systemic lupus erythematosus (SLE). Herein, we summarize earlier work on epigenetic mechanisms contributing to human SLE. We first focus on the roles of DNA demethylation and DNA methyltransferase enzyme dysregulation, and we then review recent and important advances in this field. RECENT FINDINGS Many advances in the past year have been made. The importance of DNA demethylation in SLE was confirmed through twin studies. New T lymphocyte immune genes that are activated by DNA demethylation, and that may participate in autoreactivity, were identified. Finally, novel mechanisms contributing to DNA demethylation in SLE were discovered. SUMMARY A comprehensive understanding of the epigenetic mechanisms contributing to SLE will likely enable development of new therapeutic agents and strategies that target the dysregulated genes or correct the aberrant epigenetic modifications. Although specific agents have not yet been tested in SLE, the studies reviewed hold promise that these approaches will be useful in the treatment of human lupus.
Collapse
|
31
|
Nagy G, Koncz A, Telarico T, Fernandez D, Ersek B, Buzás E, Perl A. Central role of nitric oxide in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus. Arthritis Res Ther 2010; 12:210. [PMID: 20609263 PMCID: PMC2911902 DOI: 10.1186/ar3045] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Nitric oxide (NO) has been shown to regulate T cell functions under physiological conditions, but overproduction of NO may contribute to T lymphocyte dysfunction. NO-dependent tissue injury has been implicated in a variety of rheumatic diseases, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Several studies reported increased endogenous NO synthesis in both SLE and RA, and recent evidence suggests that NO contributes to T cell dysfunction in both autoimmune diseases. The depletion of intracellular glutathione may be a key factor predisposing patients with SLE to mitochondrial dysfunction, characterized by mitochondrial hyperpolarization, ATP depletion and predisposition to death by necrosis. Thus, changes in glutathione metabolism may influence the effect of increased NO production in the pathogenesis of autoimmunity.
Collapse
Affiliation(s)
- György Nagy
- Department of Rheumatology, Semmelweis University, Medical School, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
32
|
Smith TJ. Insulin-like growth factor-I regulation of immune function: a potential therapeutic target in autoimmune diseases? Pharmacol Rev 2010; 62:199-236. [PMID: 20392809 PMCID: PMC2879913 DOI: 10.1124/pr.109.002469] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This topically limited review explores the relationship between the immune system and insulin-like growth factors (IGF-I and IGF-II) and the proteins through which they act, including IGF-I receptor (IGF-IR) and the IGF-I binding proteins. The IGF/IGF-IR pathway plays important and diverse roles in tissue development and function. It regulates cell cycle progression, apoptosis, and the translation of proteins. Many of the consequences ascribed to IGF-IR activation result from its association with several accessory proteins that are either identical or closely related to those involved in insulin receptor signaling. Relatively recent awareness that IGF-I and IGF-IR regulate immune function has cast this pathway in an unexpected light; it may represent an important switch governing the quality and amplitude of immune responses. IGF-I/IGF-IR signaling may also participate in the pathogenesis of autoimmune diseases, although its relationship with these processes seems complex and relatively unexplored. On the one hand, IGF-I seems to protect experimental animals from developing insulin-deficient diabetes mellitus. In contrast, activating antibodies directed at IGF-IR have been detected in patients with Graves' disease, where the receptor is overexpressed by multiple cell types. The frequency of IGF-IR+ B and T cells is substantially increased in patients with that disease. Potential involvement of IGF-I and IGF-IR in the pathogenesis of autoimmune diseases suggests that this pathway might constitute an attractive therapeutic target. IGF-IR has been targeted in efforts directed toward drug development for cancer, employing both small-molecule and monoclonal antibody approaches. These have been generally well-tolerated. Recognizing the broader role of IGF-IR in regulating both normal and pathological immune responses may offer important opportunities for therapeutic intervention in several allied diseases that have proven particularly difficult to treat.
Collapse
Affiliation(s)
- Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| |
Collapse
|
33
|
Via CS, Shearer GM. Defective in vitro IL-2 production in lupus is an early but secondary event paralleling disease activity: evidence from the murine parent-into-F1 model supports staging of IL-2 defects in human lupus. Autoimmunity 2010; 43:23-31. [PMID: 20001649 DOI: 10.3109/08916930903374808] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cell defects are a well described feature of both human and murine lupus however their exact significance is unclear. Evidence from an induced model of lupus, the P --> F1 model of chronic lupus-like GVHD demonstrates that a secondary inducible T cell defect in in vitro IL-2 and CTL responses occurs early in the course of lupus-like disease and well in advance of clinical disease. Defective Th cell function was probed using a novel approach categorizing the response to two stimuli:1) the MHC self restricted response, termed self +X; and 2) the allogeneic response. Using this approach, lupus mice exhibited similar in vitro Th cell pattern i.e. an absent S+X response but preserved allogeneic (termed -/+). In contrast, human lupus patients exhibited three possible response patterns, +/+, - /+ or -/- with more severe in vitro T cell impairment correlated with more severe disease. Similarly, patients with other T cell mediated conditions i.e. HIV infection or renal allograft recipients, also exhibited more severe in vitro T cell impairment with greater disease activity or greater immunosuppression respectively. The similar Th response patterns in human and murine T cell mediated conditions indicates that the underlying mechanisms involved are not disease specific but instead reflect common immune responses and validate the use of the P --> F1 model for future studies of T cell mediated conditions. These results support the use of prospective monitoring of IL-2 responses in lupus patients. Successful adaptation of this approach to the clinical setting could allow not only earlier therapeutic intervention and reduced organ damage but also earlier tapering of pharmacological agents and reduced untoward effects.
Collapse
Affiliation(s)
- Charles S Via
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20841, USA.
| | | |
Collapse
|
34
|
Stanford SM, Mustelin TM, Bottini N. Lymphoid tyrosine phosphatase and autoimmunity: human genetics rediscovers tyrosine phosphatases. Semin Immunopathol 2010; 32:127-36. [PMID: 20204370 DOI: 10.1007/s00281-010-0201-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 01/28/2010] [Indexed: 01/22/2023]
Abstract
A relatively large number of protein tyrosine phosphatases (PTPs) are known to regulate signaling through the T cell receptor (TCR). Recent human genetics studies have shown that several of these PTPs are encoded by major autoimmunity genes. Here, we will focus on the lymphoid tyrosine phosphatase (LYP), a critical negative modulator of TCR signaling encoded by the PTPN22 gene. The functional analysis of autoimmune-associated PTPN22 genetic variants suggests that genetic variability of TCR signal transduction contributes to the pathogenesis of autoimmunity in humans.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
35
|
Jury EC, Flores-Borja F, Kalsi HS, Lazarus M, Isenberg DA, Mauri C, Ehrenstein MR. Abnormal CTLA-4 function in T cells from patients with systemic lupus erythematosus. Eur J Immunol 2010; 40:569-78. [PMID: 19950182 DOI: 10.1002/eji.200939781] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CTLA-4 is a critical gatekeeper of T-cell activation and immunological tolerance and has been implicated in patients with a variety of autoimmune diseases through genetic association. Since T cells from patients with the autoimmune disease systemic lupus erythematosus (SLE) display a characteristic hyperactive phenotype, we investigated the function of CTLA-4 in SLE. Our results reveal increased CTLA-4 expression in FOXP3(-) responder T cells from patients with SLE compared with other autoimmune rheumatic diseases and healthy controls. However, CTLA-4 was unable to regulate T-cell proliferation, lipid microdomain formation and phosphorylation of TCR-zeta following CD3/CD28 co-stimulation, in contrast to healthy T cells. Although lupus T cells responded in vitro to CD3/CD28 co-stimulation, there was no parallel increase in CTLA-4 expression, which would normally provide a break on T-cell proliferation. These defects were associated with exclusion of CTLA-4 from lipid microdomains providing an anatomical basis for its loss of function. Collectively our data identify CTLA-4 dysfunction as a potential cause for abnormal T-cell activation in patients with SLE, which could be targeted for therapy.
Collapse
Affiliation(s)
- Elizabeth C Jury
- Department of Medicine, Centre for Rheumatology, University College London, London, UK.
| | | | | | | | | | | | | |
Collapse
|
36
|
Crispín JC, Liossis SNC, Kis-Toth K, Lieberman LA, Kyttaris VC, Juang YT, Tsokos GC. Pathogenesis of human systemic lupus erythematosus: recent advances. Trends Mol Med 2010; 16:47-57. [PMID: 20138006 DOI: 10.1016/j.molmed.2009.12.005] [Citation(s) in RCA: 266] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 12/21/2009] [Accepted: 12/21/2009] [Indexed: 01/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with manifestations derived from the involvement of multiple organs including the kidneys, joints, nervous system and hematopoietic organs. Immune system aberrations, as well as heritable, hormonal and environmental factors interplay in the expression of organ damage. Recent contributions from different fields have developed our understanding of SLE and reshaped current pathogenic models. Here, we review recent findings that deal with (i) genes associated with disease expression; (ii) immune cell molecular abnormalities that lead to autoimmune pathology; (iii) the role of hormones and sex chromosomes in the development of disease; and (iv) environmental and epigenetic factors thought to contribute to the expression of SLE. Finally, we highlight molecular defects intimately associated with the disease process of SLE that might represent ideal therapeutic targets and disease biomarkers.
Collapse
Affiliation(s)
- José C Crispín
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Ho JWK, Lin MW, Braet F, Su YY, Adelstein S, dos Remedios CG. Customising an antibody leukocyte capture microarray for systemic lupus erythematosus: beyond biomarker discovery. Proteomics Clin Appl 2009; 4:179-89. [PMID: 21137042 DOI: 10.1002/prca.200900165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 08/10/2009] [Accepted: 09/17/2009] [Indexed: 11/10/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that has heterogeneous clinical manifestation with diverse patterns of organ involvement, autoantibody profiles and varying degrees of severity of disease. Research and clinical experience indicate that different subtypes of SLE patients will likely benefit from more tailored treatment regimes, but we currently lack a fast and objective test with high enough sensitivity to enable us to perform such sub-grouping for clinical use. In this article, we review how proteomic technologies could be used as such an objective test. In particular, we extensively review many leukocyte surface markers that are known to have an association with the pathogenesis of SLE, and we discuss how these markers can be used in the further development of a novel SLE-specific antibody leukocyte capture microarray. In addition, we review some bioinformatics challenges and current methods for using the data generated by these cell-capture microarrays in clinical use. In a broader context, we hope our experience in developing a disease specific cell-capture microarray for clinical application can be a guide to other proteomic practitioners who intend to extend their technologies to develop clinical diagnostic and prognostic tests for complex diseases.
Collapse
Affiliation(s)
- Joshua W K Ho
- Muscle Research Unit, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
38
|
Nicolaou SA, Neumeier L, Takimoto K, Lee SM, Duncan HJ, Kant SK, Mongey AB, Filipovich AH, Conforti L. Differential calcium signaling and Kv1.3 trafficking to the immunological synapse in systemic lupus erythematosus. Cell Calcium 2009; 47:19-28. [PMID: 19959227 DOI: 10.1016/j.ceca.2009.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 10/27/2009] [Accepted: 11/02/2009] [Indexed: 11/24/2022]
Abstract
Systemic lupus erythematosus (SLE) T cells exhibit several activation signaling anomalies including defective Ca(2+) response and increased NF-AT nuclear translocation. The duration of the Ca(2+) signal is critical in the activation of specific transcription factors and a sustained Ca(2+) response activates NF-AT. Yet, the distribution of Ca(2+) responses in SLE T cells is not known. Furthermore, the mechanisms responsible for Ca(2+) alterations are not fully understood. Kv1.3 channels control Ca(2+) homeostasis in T cells. We reported a defect in Kv1.3 trafficking to the immunological synapse (IS) of SLE T cells that might contribute to the Ca(2+) defect. The present study compares single T cell quantitative Ca(2+) responses upon formation of the IS in SLE, normal, and rheumatoid arthritis (RA) donors. Also, we correlated cytosolic Ca(2+) concentrations and Kv1.3 trafficking in the IS by two-photon microscopy. We found that sustained [Ca(2+)](i) elevations constitute the predominant response to antigen stimulation of SLE T cells. This defect is selective to SLE as it was not observed in RA T cells. Further, we observed that in normal T cells termination of Ca(2+) influx is accompanied by Kv1.3 permanence in the IS, while Kv1.3 premature exit from the IS correlates with sustained Ca(2+) responses in SLE T cells. Thus, we propose that Kv1.3 trafficking abnormalities contribute to the altered distribution in Ca(2+) signaling in SLE T cells. Overall these defects may explain in part the T cell hyperactivity and dysfunction documented in SLE patients.
Collapse
Affiliation(s)
- Stella A Nicolaou
- Department of Internal Medicine, University of Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Translational control is an important but relatively unappreciated mechanism that regulates levels of protein products. In addition to a global translational control that regulates the cell's response to external stimuli such as growth factors, cytokines, stress, and viral infections, selective translational control has recently been demonstrated to affect many genes related to growth and apoptotic processes. Translational infidelity has recently been suggested as a new mechanism of T cell dysregulation in SLE. This review discusses current data on translational control of T cell biology and the central aspect of translational control in the signalling pathway leading to T cell proliferation, apoptotic response, and cytokine production. The utility for global analysis by genomics to study translational control of T cell gene expression is also discussed.
Collapse
Affiliation(s)
- Laura Beretta
- Department of Microbiology and Immunology, University of Michigan, Medical School, Ann Arbor, 48109-0620, USA.
| |
Collapse
|
40
|
Kammer GM, Laxminarayana D, Khan IU. MECHANISMS OF DEFICIENT TYPE I PROTEIN KINASE A ACTIVITY IN LUPUS T LYMPHOCYTES. Int Rev Immunol 2009; 23:225-44. [PMID: 15204086 DOI: 10.1080/08830180490452611] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which the immune response to antigen results in exaggerated CD4(+) T helper and diminished CD8(+) T cytotoxic responses. To determine the mechanisms underlying impaired T cell effector functions, we have investigated the cAMP/protein kinase A (cAMP/PKA) signaling pathway. The results demonstrate that diminished PKA-catalyzed protein phosphorylation is the result of deficient type I (PKA-I) and type II (PKA-II) isozyme-specific activities. The prevalence of deficient PKA-I and PKA-II activities in SLE T cells is approximately 80% and 40%, respectively. Diminished PKA-I activities are not associated with disease activity and appear to be stable over time. Two disparate mechanisms account for these low PKA-I and PKA-II isozyme activities. Moreover, novel transcript mutations of the RI alpha gene have been identified that are characterized by deletions, transitions, and transversions. Most mutations are clustered adjacent to GAGAG motifs and CT repeats. In conclusion, aberrant signaling via the cAMP/PKA pathway occurs in SLE T cells, and this is proposed to contribute to abnormal T cell effector functions.
Collapse
Affiliation(s)
- Gary M Kammer
- Section on Rheumatology and Clinical Immunology, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | |
Collapse
|
41
|
BLASINI ANAM, RODRíGUEZ MARTA. ALTERED SIGNALING TRIGGERED BY LIGATION OF THE TCR/CD3 RECEPTOR IN T LYMPHOCYTES FROM PATIENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS: THE ROAD FROM ANERGY TO AUTOIMMUNITY. Int Rev Immunol 2009. [DOI: 10.1080/08830180490452594-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease, the exact etiology of which is unknown. It is characterized by the production of pathological autoantibodies which adhere to cellular surfaces or form immune complexes which deposit in tissue, leading to end-organ damage via inflammatory mechanisms including complement activation. SLE may manifest itself in any organ system. In the eye, keratoconjunctivitis sicca is the most common finding. Other ophthalmic sites of involvement include the cornea, conjunctiva, episclera, sclera, uveal tract, retina, vasculature, optic nerve, and orbit. Therapy varies based on the disease manifestation and severity.
Collapse
Affiliation(s)
- Russell W Read
- Department of Ophthalmology and Pathology, University of Alabama at Birmingham, AL 35233, USA.
| |
Collapse
|
43
|
Takeuchi T, Tsuzaka K, Abe T, Yoshimoto K, Shiraishi K, Kameda H, Amano K. T cell abnormalities in systemic lupus erythematosus. Autoimmunity 2009; 38:339-46. [PMID: 16227148 DOI: 10.1080/08916930500123983] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Because of the consensus that T cells play a central role in the pathogenesis of systemic lupus erythematosus (SLE), we explored the molecular basis of the defective function of SLE T cells for expression of signal transduction molecules, as well as surface structures such as adhesion molecules, by extensively testing peripheral blood T cells from SLE patients. Upregulated expression and function of adhesion molecules was observed in T cells from patients with active SLE who had specific clinical manifestations such as vasculitis, epithelitis and arthritis, but proximal signal transduction was defective. Comprehensive analysis to identify the molecules responsible for the defects showed the expression of the TCR zeta chain was attenuated, or absent in more than half of SLE patients. Moreover, the aberrant transcripts of the TCR zeta chain, including spliced variants lacking exon 7 and with a short 3' UTR, were detected in SLE T cells. Although attenuated expression of the TCR zeta chain is also observed in patients with cancers, infections and other autoimmune diseases, sustained attenuation of TCR zeta expression and aberrant transcripts are only observed in SLE. In this review we discuss the unique features of the TCR zeta defects in SLE.
Collapse
|
44
|
Huang YC, Hung WC, Chen WT, Yu HS, Chai CY. Sodium arsenite-induced DAPK promoter hypermethylation and autophagy via ERK1/2 phosphorylation in human uroepithelial cells. Chem Biol Interact 2009; 181:254-62. [PMID: 19577553 DOI: 10.1016/j.cbi.2009.06.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 06/16/2009] [Accepted: 06/25/2009] [Indexed: 12/19/2022]
Abstract
Arsenic compounds or arsenicals are well-known toxic and carcinogenic agents. The toxic effects of arsenic that are of most concern to humans are those that occur from chronic, low-level exposure, and are associated with various human malignancies, including skin, lung and bladder cancers. In addition, arsenic could induce cell death, including apoptosis or autophagy in malignant cells. Previously, we have demonstrated that arsenite can induce autophagy and death-associated protein kinase (DAPK) promoter hypermethylation in the SV-40 immortalized human uroepithelial cell line (SV-HUC-1). However, the underlying mechanism of arsenite-induced autophagy is still unclear. In the present study, we demonstrate that arsenite can activate the extracellular signaling-regulated protein kinase 1/2 (ERK1/2) signaling pathway after treatment in SV-HUC-1 cells by using immunocytochemistry and Western blotting. In addition, our results also show an increase of autophagosomes was produced in arsenite-treated SV-HUC-1 cells by using electron microscopy. We found that, by incrementally increasing the dosages, microtubule-associated protein light chain 3B (LC3B) and Beclin-1 are important regulators for the formation of autophagosomes, in a dose-dependent manner. When the cells were pretreated with inhibitors 5-aza-CdR or U0126 for 24h, the effect of arsenite on ERK1/2, LC3B, Beclin-1 and DAPK proteins expression is suppressed. Furthermore, our results support the notion that arsenite can induce the ERK1/2 signaling pathway to stimulate autophagy and DAPK promoter hypermethylation in human uroepithelial SV-HUC-1 cells. These findings may contribute to a better understanding of the carcinogenesis of arsenite.
Collapse
|
45
|
Sunahori K, Juang YT, Tsokos GC. Methylation status of CpG islands flanking a cAMP response element motif on the protein phosphatase 2Ac alpha promoter determines CREB binding and activity. THE JOURNAL OF IMMUNOLOGY 2009; 182:1500-8. [PMID: 19155497 DOI: 10.4049/jimmunol.182.3.1500] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine protein phosphatase in eukaryotic cells and is involved in many essential aspects of cell function. The catalytic subunit of the enzyme (PP2Ac), a part of the core enzyme, has two isoforms, alpha (PP2Ac alpha) and beta (PP2Ac beta), of which PP2Ac alpha is the major form expressed in vivo. Deregulation of PP2A expression has been linked to several diseases, but the mechanisms that control the expression of this enzyme are still unclear. We conducted experiments to decipher molecular mechanisms involved in the regulation of the PP2Ac alpha promoter in human primary T cells. After preparing serially truncated PP2Ac alpha promoter luciferase constructs, we found that the region stretching around 240 bases upstream from the translation initiation site was of functional significance and included a cAMP response element motif flanked by three GC boxes. Shift assays revealed that CREB/phosphorylated CREB and stable protein 1 could bind to the region. Furthermore, we demonstrated that methylation of deoxycytosine in the CpG islands limited binding of phosphorylated CREB and the activity of the PP2Ac alpha promoter. In contrast, the binding of stable protein 1 to a GC box within the core promoter region was not affected by DNA methylation. Primary T cells treated with 5-azacitidine, a DNA methyltransferase inhibitor, showed increased expression of PP2Ac alpha mRNA. We propose that conditions associated with hypomethylation of CpG islands, such as drug-induced lupus, permit increased PP2Ac expression.
Collapse
Affiliation(s)
- Katsue Sunahori
- Division of Rheumatology in Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
46
|
Kong KO, Tan AW, Thong BYH, Lian TY, Cheng YK, Teh CL, Koh ET, Chng HH, Law WG, Lau TC, Leong KP, Leung BP, Howe HS. Enhanced expression of interferon-inducible protein-10 correlates with disease activity and clinical manifestations in systemic lupus erythematosus. Clin Exp Immunol 2009; 156:134-40. [PMID: 19175619 DOI: 10.1111/j.1365-2249.2009.03880.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Our objective was to investigate the serum levels of interferon-inducible protein-10 (IP-10) in systemic lupus erythematosus (SLE) and their correlation with disease activity and organ manifestations. Serum IP-10 levels were assessed in 464 SLE patients and 50 healthy donors. Disease activity was assessed by the revised SLE Activity Measure, and the concomitant active organ manifestations, anti-ds DNA antibody titres, complement levels and erythrocyte sedimentation rates recorded. Peripheral blood mononuclear cell (PBMC) synthesis of IP-10 in SLE patients and controls was determined by in vitro cultures stimulated with mitogen or lipopolysaccharide. Elevated serum IP-10 levels were observed in SLE patients, which were significantly higher in the presence of active haematological and mucocutaneous manifestations. SLE PBMCs exhibited enhanced spontaneous IP-10 production in vitro. Serial IP-10 levels correlated with longitudinal change in SLE activity, even at low levels where anti-dsDNA antibody and complement levels remain unchanged. These data demonstrate that IP-10 levels are increased in SLE and serum IP-10 may represent a more sensitive marker for monitoring disease activity than standard serological tests.
Collapse
Affiliation(s)
- K O Kong
- Department of Rheumatology, Allergy and Immunology, Tan Tock Seng Hospital, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Krishnan S, Juang YT, Chowdhury B, Magilavy A, Fisher CU, Nguyen H, Nambiar MP, Kyttaris V, Weinstein A, Bahjat R, Pine P, Rus V, Tsokos GC. Differential expression and molecular associations of Syk in systemic lupus erythematosus T cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:8145-52. [PMID: 19018007 DOI: 10.4049/jimmunol.181.11.8145] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diminished expression of TCR zeta and reciprocal up-regulation and association of FcRgamma with the TCR/CD3 complex is a hallmark of systemic lupus erythematosus (SLE) T cells. In this study we explored whether differential molecular associations of the spleen tyrosine kinase Syk that preferentially binds to FcRgamma contribute to pathological amplification of signals downstream of this "rewired TCR" in SLE. We detected higher amounts of Syk expression and activity in SLE compared with normal T cells. Selective inhibition of the activity of Syk reduced the strength of TCR-induced calcium responses and slowed the rapid kinetics of actin polymerization exclusively in SLE T cells. Syk and ZAP-70 also associated differently with key molecules involved in cytoskeletal and calcium signaling in SLE T cells. Thus, while Vav-1 and LAT preferentially bound to Syk, phospholipase C-gamma1 bound to both Syk and ZAP-70. Our results show that differential associations of Syk family kinases contribute to the enhanced TCR-induced signaling responses in SLE T cells. Thus, we propose molecular targeting of Syk as a measure to control abnormal T cell responses in SLE.
Collapse
Affiliation(s)
- Sandeep Krishnan
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lu MC, Lai NS, Yu HC, Hsieh SC, Tung CH, Yu CL. Nifedipine suppresses Th1/Th2 cytokine production and increased apoptosis of anti-CD3 + anti-CD28-activated mononuclear cells from patients with systemic lupus erythematosus via calcineurin pathway. Clin Immunol 2008; 129:462-70. [DOI: 10.1016/j.clim.2008.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 07/26/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
|
49
|
Gorjestani S, Rider V, Kimler BF, Greenwell C, Abdou NI. Extracellular signal-regulated kinase 1/2 signalling in SLE T cells is influenced by oestrogen and disease activity. Lupus 2008; 17:548-54. [PMID: 18539708 DOI: 10.1177/0961203307087982] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that occurs primarily in women of reproductive age. The disease is characterized by exaggerated T-cell activity and abnormal T-cell signalling. The mitogen-activated protein kinase (MAPK) pathway is involved in the maintenance of T-cell tolerance that fails in patients with SLE. Oestrogen is a female sex hormone that binds to nuclear receptors and alters the rate of gene transcription. Oestrogen can also act through the plasma membrane and rapidly stimulate second messengers including calcium flux and kinase activation. In this study, we investigated whether oestrogen influences the activation of MAPK signalling through the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in activated SLE T cells. SLE and control T cells were cultured in serum-free medium without and with oestradiol (10(-7) M) for 18 h. The T cells were activated with phorbol 12 myristate 13-acetate and ionomycin for various time points (0-60 min), and the amount of phosphorylated ERK1/2 was measured by immunoblotting. There were no differences in ERK1/2 phosphorylation between SLE and control T cells at 5 and 15 min after the activation stimulus. However, comparison between the amount of phosphorylated ERK1/2 in SLE T cells from the same patients cultured without and with oestradiol showed a significant oestrogen-dependent suppression (P=0.48) of ERK1/2 in patients with inactive/mild systemic lupus erythematosus disease activity index (SLEDAI) (0-2) compared with patients with moderate (4-6) or active (8-12) SLEDAI scores. These results suggest that the suppression of MAPK through ERK1/2 phosphorylation is sensitive to oestradiol in patients with inactive or mild disease, but the sensitivity is not maintained when disease activity increases. Furthermore, studies are now necessary to understand the mechanisms by which oestrogen influences MAPK activation in SLE T cells.
Collapse
Affiliation(s)
- S Gorjestani
- Department of Biology, Pittsburg State University, Pittsburg, Kansas 66762, USA
| | | | | | | | | |
Collapse
|
50
|
Gorelik G, Richardson B. Aberrant T cell ERK pathway signaling and chromatin structure in lupus. Autoimmun Rev 2008; 8:196-8. [PMID: 18723128 DOI: 10.1016/j.autrev.2008.07.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibodies to nuclear components with subsequent immune complex formation and deposition in multiple organs. A combination of genetic and environmental factors is required for disease development, but how the environment interacts with the immune system in genetically predisposed hosts to cause lupus is unclear. Recent evidence suggests that environmental agents may alter T cell chromatin structure and gene expression through effects on DNA methylation, a repressive epigenetic mechanism promoting chromatin inactivation, to cause lupus in people with the appropriate genetic background. DNA methylation is regulated by ERK pathway signaling, and abnormalities in ERK pathway signaling may contribute to immune dysfunction in lupus through epigenetic effects on gene expression. This article reviews current evidence for epigenetic abnormalities, and in particular DNA demethylation, in the pathogenesis of idiopathic and some forms of drug-induced lupus, and how impaired ERK pathway signaling may contribute to the development of human lupus through effects on T cell DNA methylation.
Collapse
Affiliation(s)
- Gabriela Gorelik
- Department of Medicine, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | |
Collapse
|