51
|
Tangtanatakul P, Thammasate B, Jacquet A, Reantragoon R, Pisitkun T, Avihingsanon Y, Leelahavanichkul A, Hirankarn N. Transcriptomic profiling in human mesangial cells using patient-derived lupus autoantibodies identified miR-10a as a potential regulator of IL8. Sci Rep 2017; 7:14517. [PMID: 29109423 PMCID: PMC5673966 DOI: 10.1038/s41598-017-15160-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 10/23/2017] [Indexed: 11/09/2022] Open
Abstract
Autoantibody-mediated inflammation directed at resident kidney cells mediates lupus nephritis (LN) pathogenesis. This study investigated the role of miRNA in human mesangial cells (HMCs) stimulated with auto anti-dsDNA immunoglobulin (Ig)G antibodies. HMCs were treated with antibodies purified from active LN patients or non-specific IgG controls in the presence of normal serum. Aberrant miRNA was screened using high throughput sequencing. Anti-dsDNA IgG up-regulated 103 miRNAs and down-regulated 30 miRNAs. The miRNAs regulated genes in the cell cycle, catabolic processes, regulation of transcription and apoptosis signalling. miR-10a was highly abundant in HMCs but was specifically downregulated upon anti-dsDNA IgG induction. Interestingly, the expression of miR-10a in kidney biopsies from class III and IV LN patients (n = 26) was downregulated compared with cadaveric donor kidneys (n = 6). Functional studies highlighted the downstream regulator of miR-10a in the chemokine signalling and cell proliferation or apoptosis pathways. Luciferase assay confirmed for the first time that IL8 was a direct target of miR-10a in HMCs. In conclusion, anti-dsDNA IgG Ab down-regulated miR-10a expression in HMCs resulting in the induction of various target genes involved in HMC proliferation and chemokine expression.
Collapse
Affiliation(s)
- Pattarin Tangtanatakul
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand.,Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Boonyakiat Thammasate
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Alain Jacquet
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsima Reantragoon
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Chulalongkorn University Systems Biology (CUSB), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yingyos Avihingsanon
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
52
|
Kong F, Liu Z, Jain VG, Shima K, Suzuki T, Muglia LJ, Starczynowski DT, Pasare C, Bhattacharyya S. Inhibition of IRAK1 Ubiquitination Determines Glucocorticoid Sensitivity for TLR9-Induced Inflammation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2017; 199:3654-3667. [PMID: 29038250 DOI: 10.4049/jimmunol.1700443] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/19/2017] [Indexed: 01/26/2023]
Abstract
Inflammatory responses are controlled by signaling mediators that are regulated by various posttranslational modifications. Recently, transcription-independent functions for glucocorticoids (GC) in restraining inflammation have emerged, but the underlying mechanisms are unknown. In this study, we report that GC receptor (GR)-mediated actions of GC acutely suppress TLR9-induced inflammation via inhibition of IL-1R-associated kinase 1 (IRAK1) ubiquitination. β-TrCP-IRAK1 interaction is required for K48-linked ubiquitination of IRAK1 at Lys134 and subsequent membrane-to-cytoplasm trafficking of IRAK1 interacting partners TNFR-associated factor 6 and TAK1 that facilitates NF-κB and MAPK activation. Upon costimulation of macrophages with GC and TLR9-engaging ligand, GR physically interacts with IRAK1 and interferes with protein-protein interactions between β-TrCP and IRAK1. Ablation of GR in macrophages prevents GC-dependent suppression of β-TrCP-IRAK1 interactions. This GC-mediated suppression of IRAK1 activation is unique to TLR9, as GC treatment impairs TLR9 but not TLR4 ligand-induced K48-linked IRAK1 ubiquitination and trafficking of IRAK1 interacting partners. Furthermore, mutations in IRAK1 at Lys134 prevent TLR9 ligand-induced activation of inflammatory signaling mediators and synthesis of proinflammatory cytokines to an extent comparable to GC-mediated inhibition. Collectively, these findings identify a transcription-independent, rapid, and nongenomic GC suppression of TLR9 ligand-mediated IRAK1 ubiquitination as a novel mechanism for restraining acute inflammatory reactions.
Collapse
Affiliation(s)
- Fansheng Kong
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Zhiwei Liu
- Neonatal Division, International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Viral G Jain
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Kenjiro Shima
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Takuji Suzuki
- Division of Pulmonary Biology, Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Louis J Muglia
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229
| | - Daniel T Starczynowski
- Experimental Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Chandrashekhar Pasare
- Department of Immunology, Southwestern Medical Center, University of Texas, Dallas, TX 75390
| | - Sandip Bhattacharyya
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229;
| |
Collapse
|
53
|
Phenotyping of autoreactive B cells with labeled nucleosomes in 56R transgenic mice. Sci Rep 2017; 7:13232. [PMID: 29038433 PMCID: PMC5643551 DOI: 10.1038/s41598-017-13422-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/21/2017] [Indexed: 01/22/2023] Open
Abstract
The phenotypic characterization of self-reactive B cells producing autoantibodies is one of the challenges to get further insight in the physiopathology of autoimmune diseases. We took advantage of our previously developed flow cytometry method, using labeled nucleosomes, prominent autoantigens in systemic lupus erythematosus, to analyze the phenotype of self-reactive B cells in the anti-DNA B6.56R mouse model. We showed that splenic anti-nucleosome B cells express mostly kappa light chains and harbor a marginal zone phenotype. Moreover, these autoreactive B cells fail to acquire a germinal center phenotype and are less abundant in the transitional T3 compartment. In conclusion, the direct detection of autoreactive B cells helped determine their phenotypic characteristics and provided a more direct insight into the B cell tolerance process in B6.56R mice. This method constitutes an interesting new tool to study the mechanisms of B cell tolerance breakdown in B6.56R mice crossed with autoimmune prone models.
Collapse
|
54
|
Follicular Dendritic Cell Activation by TLR Ligands Promotes Autoreactive B Cell Responses. Immunity 2017; 46:106-119. [PMID: 28099860 DOI: 10.1016/j.immuni.2016.12.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/31/2016] [Accepted: 12/06/2016] [Indexed: 12/30/2022]
Abstract
A hallmark of autoimmunity in murine models of lupus is the formation of germinal centers (GCs) in lymphoid tissues where self-reactive B cells expand and differentiate. In the host response to foreign antigens, follicular dendritic cells (FDCs) maintain GCs through the uptake and cycling of complement-opsonized immune complexes. Here, we examined whether FDCs retain self-antigens and the impact of this process in autoantibody secretion in lupus. We found that FDCs took up and retained self-immune complexes composed of ribonucleotide proteins, autoantibody, and complement. This uptake, mediated through CD21, triggered endosomal TLR7 and led to the secretion of interferon (IFN) α via an IRF5-dependent pathway. Blocking of FDC secretion of IFN-α restored B cell tolerance and reduced the amount of GCs and pathogenic autoantibody. Thus, FDCs are a critical source of the IFN-α driving autoimmunity in this lupus model. This pathway is conserved in humans, suggesting that it may be a viable therapeutic target in systemic lupus erythematosus.
Collapse
|
55
|
|
56
|
de la Varga Martínez R, Rodríguez-Bayona B, Añez GA, Medina Varo F, Pérez Venegas JJ, Brieva JA, Rodríguez C. Clinical relevance of circulating anti-ENA and anti-dsDNA secreting cells from SLE patients and their dependence on STAT-3 activation. Eur J Immunol 2017; 47:1211-1219. [DOI: 10.1002/eji.201646872] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/13/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Raquel de la Varga Martínez
- Unidad de Investigación; Hospital Universitario Puerta del Mar (HUPM); Cádiz
- Servicio de Inmunología; UGC de Hematología e Inmunología; HUPM; Cádiz
| | - Beatriz Rodríguez-Bayona
- Área de Inmunología; UGC de Análisis Clínicos; Complejo Hospitalario Universitario de Huelva; Hospital Juan Ramón Jiménez Huelva
| | - Gustavo A. Añez
- Sección de Reumatología; UGC de Cirugía Ortopédica; Traumatología y Reumatología; HUPM; Cádiz
| | - Fermín Medina Varo
- Sección de Reumatología; UGC de Cirugía Ortopédica; Traumatología y Reumatología; HUPM; Cádiz
| | | | - José A. Brieva
- Unidad de Investigación; Hospital Universitario Puerta del Mar (HUPM); Cádiz
| | - Carmen Rodríguez
- Servicio de Inmunología; UGC de Hematología e Inmunología; HUPM; Cádiz
| |
Collapse
|
57
|
Impacts of Anti-dsDNA Antibody on In Vitro Fertilization-Embryo Transfer and Frozen-Thawed Embryo Transfer. J Immunol Res 2017; 2017:8596181. [PMID: 28540311 PMCID: PMC5429951 DOI: 10.1155/2017/8596181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/27/2017] [Indexed: 11/30/2022] Open
Abstract
Our purpose is to explore whether anti-dsDNA antibody, which was demonstrated to enter living cells and induced apoptosis, could adversely affect reproductive outcomes. A total of 259 women receiving the in vitro fertilization-embryo transfer (IVF) cycle were enrolled in this study, including 52 women with positive ANA and anti-dsDNA (ANA+/anti-dsDNA+ group), 86 women with positive ANA and negative anti-dsDNA (ANA+/anti-dsDNA− group), and 121 women with negative ANA and anti-dsDNA (ANA−/anti-dsDNA− group). 136 nonpregnant women among 259 patients in the IVF-ET cycle were enrolled in the hormone replacement therapy frozen-thawed embryo transfer (HRT-TET) cycle. We compared basic characters and IVF outcomes among three groups in fresh embryo transfer and frozen-thawed embryo transfer cycle, respectively. The number of retrieved oocytes, available embryos, and high-quality embryos in the ANA+/anti-dsDNA+ group was lower than those in the other two groups in the fresh embryo transfer cycle. The rates of fertilization, implantation, and clinical pregnancy in the ANA+/anti-dsDNA+ group were the lowest, while the early miscarriage rate was the highest in the ANA+/anti-dsDNA+ group both in the fresh embryo transfer cycle and in the frozen-thawed embryo transfer cycle. Our data suggested that anti-dsDNA antibody may be the essential marker for defective oocytes or embryos in infertile women with any type of ANA.
Collapse
|
58
|
Hoyer BF, Radbruch A. Protective and pathogenic memory plasma cells. Immunol Lett 2017; 189:10-12. [PMID: 28455167 DOI: 10.1016/j.imlet.2017.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
The immune system can be divided into two major parts: innate and adaptive immunity. Adaptive immunity is characterized by its major cellular players: the B and T cells. B cells will, in the context of an immune reaction, differentiate into plasma cells. These plasma cells produce antibodies, which are secreted. Antibodies are characterized by their specificity against a selected antigen and by their isotype. The isotype changes with the duration or phase of the immune reaction. Early immune reactions are usually characterized by the predominant production of IgM antibodies. With the persistence of the immune reaction immunoglobulin class switch occurs and plasma cells will produce IgG, IgE or IgA-antibodies (Radbruch et al., 2006) [1].
Collapse
Affiliation(s)
- Bimba F Hoyer
- Charité Universitätsmedizin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum-a Leibniz Institute Berlin, Germany.
| | - Andreas Radbruch
- Charité Universitätsmedizin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum-a Leibniz Institute Berlin, Germany
| |
Collapse
|
59
|
Identification of autoreactive B cells with labeled nucleosomes. Sci Rep 2017; 7:602. [PMID: 28377609 PMCID: PMC5428865 DOI: 10.1038/s41598-017-00664-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/28/2017] [Indexed: 12/15/2022] Open
Abstract
The pathogenesis of autoimmune diseases has not been completely elucidated yet, and only a few specific treatments have been developed so far. In autoimmune diseases mediated by pathogenic autoantibodies, such as systemic lupus erythematosus, the specific detection and analysis of autoreactive B cells is crucial for a better understanding of the physiopathology. Biological characterization of these cells may help to define new therapeutic targets. Very few techniques allowing the precise detection of autoreactive B cells have been described so far. Herein we propose a new flow cytometry technique for specific detection of anti-nucleosome B cells, which secrete autoantibodies in systemic lupus erythematosus, using labeled nucleosomes. We produced different fluorochrome-labeled nucleosomes, characterized them, and finally tested them in flow cytometry. Nucleosomes labeled via the cysteines present in H3 histone specifically bind to autoreactive B cells in the anti-DNA transgenic B6.56R mice model. The present work validates the use of fluorochrome-labeled nucleosomes via cysteines to identify anti-nucleosome B cells and offers new opportunities for the description of autoreactive B cell phenotype.
Collapse
|
60
|
Yung S, Chan TM. Molecular and Immunological Basis of Tubulo-Interstitial Injury in Lupus Nephritis: a Comprehensive Review. Clin Rev Allergy Immunol 2017; 52:149-163. [PMID: 26961386 DOI: 10.1007/s12016-016-8533-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lupus nephritis is an important cause of kidney failure in patients of Asian, African, or Hispanic descent. Its etiology and pathogenesis are multifactorial and remain to be elucidated. Accumulating evidence suggests that anti-double-stranded DNA (dsDNA) antibodies play a critical role in the pathogenesis, through its direct binding to cross-reactive antigens on resident renal cells or indirect binding through chromatin material to extracellular matrix components, resulting in complement activation, cell activation and proliferation, and induction of inflammatory and fibrotic processes. While tubulo-interstitial damage portends poor long-term renal prognosis, the mechanisms leading to tubulo-interstitial injury in lupus nephritis has received relatively less attention to date. Immune deposition along the tubular basement membrane is often observed in lupus nephritis and correlates with tubulo-interstitial infiltration of immune cells and interstitial fibrosis. Anti-dsDNA antibodies bind to resident renal cells, including proximal renal tubular epithelial cells, and contribute to renal inflammation and fibrosis. There is emerging evidence that epigenetic influence such as DNA methylation, histone modification, and microRNAs (miRs) also contribute to kidney fibrosis. Overexpression of miR-150 is observed in renal biopsies from patients with lupus nephritis and correlates with kidney fibrosis and chronicity score. Mycophenolate mofetil (MMF) is an established and effective standard-of-care therapy for patients with lupus nephritis. Accumulating data suggest that in addition to its immunosuppressive actions on lymphocyte proliferation, mycophenolic acid (MPA), the active metabolite of MMF, can exert a direct effect on nonimmune cells. Mediators of inflammation and fibrosis induced by anti-dsDNA antibodies in cultured proximal renal tubular epithelial cells are ameliorated by the addition of MPA, suggesting that in addition to its immunosuppressive actions, MPA may also have a beneficial effect in improving tubulo-interstitial inflammation and fibrosis through its direct action on proximal renal tubular epithelial cells.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Tak Mao Chan
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
61
|
Luo Q, Ye J, Zeng L, Li X, Fang L, Ju B, Huang Z, Li J. Elevated expression of TIGIT on CD3 +CD4 + T cells correlates with disease activity in systemic lupus erythematosus. Allergy Asthma Clin Immunol 2017; 13:15. [PMID: 28261278 PMCID: PMC5331638 DOI: 10.1186/s13223-017-0188-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 02/18/2017] [Indexed: 01/27/2023] Open
Abstract
Objectives It is well-known that lymphocytes play an important role in systemic lupus erythematosus (SLE). T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory domains (TIGIT) is one of immunosuppressive costimulatory molecules that mediates an inhibitory effect. However, its roles in SLE are poorly understood. This study was designed to investigate the correlation between the frequencies of TIGIT-expressing CD3+CD4+ T lymphocytes and SLE. Methods Patients with SLE were recruited from the First Affiliated Hospital of Nanchang University. Medical history, clinical manifestations, physical examination and laboratory measurements were recorded. The expression of TIGIT on CD3+ T lymphocytes, B lymphocytes, monocytes, neutrophils, CD3+CD4+ T lymphocytes and CD3+CD8+ T lymphocytes were determined by flow cytometry. The frequencies of TIGIT-expressing CD3+CD4+ T lymphocytes in patients with SLE were further analyzed for correlations with markers of autoimmune response, inflammation, urine proteins and disease activity in SLE. Results The frequency of TIGIT-expressing CD3+CD4+ T lymphocytes was significantly elevated in SLE patients compared with healthy controls (P < 0.0001). The frequency of TIGIT-expressing CD3+CD4+ T lymphocytes in patients with SLE was increased significantly in subjects with high anti-dsDNA titer (P = 0.026), high anti-Sm titer (P = 0.026), and high levels of urine microalbumin (P = 0.046). Furthermore, The frequency of TIGIT-expressing CD3+CD4+ T lymphocytes was found to be positively correlated with the Disease Activity Index (SLEDAI) score in SLE (r2 = 0.082; P = 0.044). Conclusion In SLE, the frequency of TIGIT-expressing CD3+CD4+ T lymphocytes was elevated and associated with the disease activity.
Collapse
Affiliation(s)
- Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Jianqing Ye
- Department of Medical College, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Lulu Zeng
- Department of Medical College, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Xue Li
- Department of Medical College, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Le Fang
- Department of Blood Transfusion, 521 Hospital of Ordnance Industry, Xi'an, 710065 Shanxi China
| | - Beihua Ju
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Zikun Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| |
Collapse
|
62
|
Annexin II-binding immunoglobulins in patients with lupus nephritis and their correlation with disease manifestations. Clin Sci (Lond) 2017; 131:653-671. [PMID: 28183811 DOI: 10.1042/cs20160732] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 01/07/2023]
Abstract
Annexin II on mesangial cell surface mediates the binding of anti-dsDNA antibodies and consequent downstream inflammatory and fibrotic processes. We investigated the clinical relevance of circulating annexin II-binding immunoglobulins (Igs) in patients with severe proliferative lupus nephritis, and renal annexin II expression in relation to progression of nephritis in New Zealand Black and White F1 mice (NZBWF1/J) mice. Annexin II-binding Igs in serum were measured by ELISA. Ultrastructural localization of annexin II was determined by electron microscopy. Seropositivity rates for annexin II-binding IgG and IgM in patients with active lupus nephritis were significantly higher compared with controls (8.9%, 1.3% and 0.9% for annexin II-binding IgG and 11.1%, 4.0% and 1.9% for annexin II-binding IgM for patients with active lupus nephritis, patients with non-lupus renal disease and healthy subjects respectively). In lupus patients, annexin II-binding IgM level was higher at disease flare compared with remission. Annexin II-binding IgG and IgM levels were associated with that of anti-dsDNA and disease activity. Annexin II-binding IgG and IgM levels correlated with histological activity index in lupus nephritis biopsy samples. In NZBWF1/J mice, serum annexin II-binding IgG and IgM levels and glomerular annexin II and p11 expression increased with progression of active nephritis. Annexin II expression was present on mesangial cell surface and in the mesangial matrix, and co-localized with electron-dense deposits along the glomerular basement membrane. Our results show that circulating annexin II-binding IgG and IgM levels are associated with clinical and histological disease activity in proliferative lupus nephritis. The co-localization of annexin II and p11 expression with immune deposition in the kidney suggests pathogenic relevance.
Collapse
|
63
|
Jang SH, Chen H, Gregersen PK, Diamond B, Kim SJ. Kruppel-like factor4 regulates PRDM1 expression through binding to an autoimmune risk allele. JCI Insight 2017; 2:e89569. [PMID: 28097234 DOI: 10.1172/jci.insight.89569] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A SNP identified as rs548234, which is found in PRDM1, the gene that encodes BLIMP1, is a risk allele associated with systemic lupus erythematosus (SLE). BLIMP1 expression was reported to be decreased in women with the PRDM1 rs548234 risk allele compared with women with the nonrisk allele in monocyte-derived DCs (MO-DCs). In this study, we demonstrate that BLIMP1 expression is regulated by the binding of Kruppel-like factor 4 (KLF4) to the risk SNP. KLF4 is highly expressed in MO-DCs but undetectable in B cells, consistent with the lack of altered expression of BLIMP1 in B cells from risk SNP carriers. Female rs548234 risk allele carriers, but not nonrisk allele carriers, exhibited decreased levels of BLIMP1 in MO-DCs, showing that the regulatory function of KLF4 is influenced by the risk allele. In addition, KLF4 directly recruits histone deacetylases (HDAC4, HDAC6, and HDAC7), established negative regulators of gene expression. Finally, the knock down of KLF4 expression reversed the inhibitory effects of the risk SNP on promoter activity and BLIMP1 expression. Therefore, the binding of KLF4 and the subsequent recruitment of HDACs represent a mechanism for reduced BLIMP1 expression in MO-DCs bearing the SLE risk allele rs548234.
Collapse
Affiliation(s)
- Su Hwa Jang
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Helen Chen
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Peter K Gregersen
- Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, New York, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Diseases and
| |
Collapse
|
64
|
Panza F, Alcaro MC, Petrelli F, Angelotti F, Pratesi F, Rovero P, Migliorini P. A novel DNA/histone H4 peptide complex detects autoantibodies in systemic lupus erythematosus sera. Arthritis Res Ther 2016; 18:220. [PMID: 27716380 PMCID: PMC5050916 DOI: 10.1186/s13075-016-1117-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/13/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The detection of anti-dsDNA antibodies is critical for the diagnosis and follow-up of systemic lupus erythematosus (SLE) patients. The presently available assays are characterized by a non-optimal specificity (solid phase assays) or sensitivity (Crithidia Luciliae immunofluorescence test (CLIFT)). To overcome the limits of CLIFT and solid phase chromatin assays, we explored the diagnostic potential of an assay based on plasmid DNA containing a highly bent fragment of 211 bp from Crithidia Luciliae minicircles, complexed with histone peptides. METHODS Electrically neutral complexes of PK201/CAT plasmid (PK) DNA and histone 4 (H4) peptides were evaluated by electromobility shift assay. Complexes of H4 peptides and PK were absorbed to the solid phase to detect specific immunoglobulin G (IgG) in sera. Sera from 109 SLE patients, 100 normal healthy subjects, and 169 disease controls were tested. RESULTS H4(14-34) containing the consensus sequence for DNA binding interacts with PK, retarding its migration. H4(14-34)/PK complexes were used to test sera by ELISA. Anti-H4-PK antibodies were detected in 56 % of SLE sera (more frequently in patients with skin or joint involvement) versus 5.9 % in disease controls; inhibition assays show that sera react with epitopes present on DNA or on the complex, not on the peptide. Antibody titer is correlated with European Consensus Lupus Activity Measurement (ECLAM) score and anti-complement component 1q (C1q) antibodies, negatively with C3 levels. Anti-H4-PK antibodies compared with CLIFT and solid phase dsDNA assays display moderate concordance. CONCLUSIONS The H4/PK assay is a simple and reliable test which is useful for the differential diagnosis and evaluation of disease activity in SLE patients.
Collapse
Affiliation(s)
- Filomena Panza
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Maria Claudia Alcaro
- Toscana Biomarkers Srl, Siena, Italy.,Present address: DIESSE Diagnostica Senese SPA, via Fiorentina 1, 53100, Siena, Italy
| | - Fiorella Petrelli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Francesca Angelotti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Federico Pratesi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Paolo Rovero
- Laboratory of Peptide and Protein Chemistry and Biology, Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical Sciences and Nutraceutics, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Paola Migliorini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy.
| |
Collapse
|
65
|
Coutant F, Miossec P. Altered dendritic cell functions in autoimmune diseases: distinct and overlapping profiles. Nat Rev Rheumatol 2016; 12:703-715. [DOI: 10.1038/nrrheum.2016.147] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
66
|
VanPatten S, Sun S, He M, Cheng KF, Altiti A, Papatheodorou A, Kowal C, Jeganathan V, Crawford JM, Bloom O, Volpe BT, Grant C, Meurice N, Coleman TR, Diamond B, Al-Abed Y. Amending HIV Drugs: A Novel Small-Molecule Approach To Target Lupus Anti-DNA Antibodies. J Med Chem 2016; 59:8859-8867. [PMID: 27603688 DOI: 10.1021/acs.jmedchem.6b00694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease that can affect numerous tissues and is characterized by the production of nuclear antigen-directed autoantibodies (e.g., anti-dsDNA). Using a combination of virtual and ELISA-based screens, we made the intriguing discovery that several HIV-protease inhibitors can function as decoy antigens to specifically inhibit the binding of anti-dsDNA antibodies to target antigens such as dsDNA and pentapeptide DWEYS. Computational modeling revealed that HIV-protease inhibitors comprised structural features present in DWEYS and predicted that analogues containing more flexible backbones would possess preferred binding characteristics. To address this, we reduced the internal amide backbone to improve flexibility, producing new small-molecule decoy antigens, which neutralize anti-dsDNA antibodies in vitro, in situ, and in vivo. Pharmacokinetic and SLE model studies demonstrated that peptidomimetic FISLE-412,1 a reduced HIV protease inhibitor analogue, was well-tolerated, altered serum reactivity to DWEYS, reduced glomeruli IgG deposition, preserved kidney histology, and delayed SLE onset in NZB/W F1 mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Christian Grant
- BioMedical Research Models, Inc. , 67 Millbrook Street, Worcester, Massachusetts 01606, United States
| | - Nathalie Meurice
- Department of Research, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | | | | | | |
Collapse
|
67
|
Anti-dsDNA antibodies and resident renal cells - Their putative roles in pathogenesis of renal lesions in lupus nephritis. Clin Immunol 2016; 185:40-50. [PMID: 27612436 DOI: 10.1016/j.clim.2016.09.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 01/19/2023]
Abstract
Lupus nephritis affects up to 70% of patients with systemic lupus erythematosus and is an important treatable cause of kidney failure. Cardinal features of lupus nephritis include loss of self-tolerance, production of autoantibodies, immune complex deposition and immune-mediated injury to the kidney, resulting in increased cell proliferation, apoptosis, and induction of inflammatory and fibrotic processes that destroy normal nephrons. The production anti-dsDNA antibodies is a cardinal feature in lupus and their level correlates with disease activity. In addition to the formation of immune complexes thereby triggering complement activation, how anti-dsDNA antibodies home to the kidney and induce pathological processes in the renal parenchyma remain to be fully elucidated. Data from our laboratory and other investigators show that the properties of anti-dsDNA antibodies vary between patients and change over time, and that anti-dsDNA antibodies could bind directly to integral cell surface molecules such as annexin II or α-actinin, or indirectly through chromatin material deposited on the cell surface. The binding of anti-dsDNA antibodies to mesangial cells and proximal renal tubular epithelial cells triggers downstream inflammatory and fibrotic pathways, which include the activation of the PKC and MAPK signaling pathways, increased secretion of pro-inflammatory cytokines and matrix protein deposition that contribute to pathological processes in the renal parenchyma.
Collapse
|
68
|
Pedersen HL, Horvei KD, Thiyagarajan D, Seredkina N, Rekvig OP. Murine and Human Lupus Nephritis: Pathogenic Mechanisms and Theoretical Strategies for Therapy. Semin Nephrol 2016; 35:427-38. [PMID: 26573545 DOI: 10.1016/j.semnephrol.2015.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lupus nephritis is one of the most serious manifestations of systemic lupus erythematosus, and represents one of the criteria implemented to classify systemic lupus erythematosus. Although studied for decades, no consensus has been reached related to the basic cellular, molecular, and immunologic mechanism(s) responsible for lupus nephritis. No causal treatments have been developed; therapy is approached mainly with nonspecific immunosuppressive medications. More detailed insight into disease mechanisms therefore is indispensable to develop new therapeutic strategies. In this review, contemporary knowledge on the pathogenic mechanisms of lupus nephritis is discussed based on recent data in murine and human lupus nephritis. Specific focus is given to the effect of anti-double-stranded DNA/antinucleosome antibodies in the kidneys and whether they bind exposed chromatin fragments in glomeruli or whether they bind inherent glomerular structures by cross-recognition. Overall, the data presented here favor the exposed chromatin model because we did not find any indication to substantiate the anti-double-stranded DNA antibody cross-reacting model. At the end of this review we present data on why chromatin fragments are expressed in the glomeruli of patients with lupus nephritis, and discuss how this knowledge can be used to direct the development of future therapies.
Collapse
Affiliation(s)
- Hege Lynum Pedersen
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | - Kjersti Daae Horvei
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Natalya Seredkina
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Ole Petter Rekvig
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway; Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
69
|
Miyagawa F, Tagaya Y, Ozato K, Asada H. Essential Requirement for IFN Regulatory Factor 7 in Autoantibody Production but Not Development of Nephritis in Murine Lupus. THE JOURNAL OF IMMUNOLOGY 2016; 197:2167-76. [DOI: 10.4049/jimmunol.1502445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 07/13/2016] [Indexed: 12/18/2022]
|
70
|
Kalunian KC. Interferon-targeted therapy in systemic lupus erythematosus: Is this an alternative to targeting B and T cells? Lupus 2016; 25:1097-101. [DOI: 10.1177/0961203316652495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical trials of investigational agents in systemic lupus erythematosus (SLE) have focused on targeting dysregulated B and T cells; however, recent translational research findings of the importance of the dysregulation of the innate immune system in SLE have led to clinical trials that target interferon. Three biologics that target type I interferons have been tested for their efficacy and safety in active SLE patients; these phase II trials have tested the hypothesis that down-regulation of interferon-regulated gene expression (the interferon signature) lessen the clinical burden of SLE. Rontalizumab, an anti-interferon-α monoclonal antibody, was studied in patients who had discontinued immunosuppressants. This study failed to show efficacy as assessed by both two outcome assessments; however, in low interferon signature patients, response was higher and corticosteroid usage was less in rontalizumab-treated patients. Sifalimumab, another anti-interferon-α monoclonal antibody, was studied in patients who remained on standard of care therapy. This study showed significantly better efficacy in patients treated with two sifalimumab dosages; significant differences were seen in the high interferon signature group. In a similar design and in a similar population as the sifalimumab study, anifrolumab, a monoclonal antibody that binds to a type I interferon receptor, was studied in patients who remained on standard of care therapy. In this study, one dosage group demonstrated efficacy and statistically significant effects were achieved in both tested dosage groups with secondary end points. Oral corticosteroid reduction to ≤7.5 mg daily was achieved in one of the tested dosage groups and organ-specific outcomes were significantly improved in that same group. For all studies, no significant differences in serious adverse effects were seen; although, herpes zoster infections were increased in sifalimumab- and anifrolumab-treated patients and influenza rates were increased in anifrolumab-treated patients. Anifrolumab is currently in pivotal phase III studies. Data appear to support the concept that targeting type I interferon in SLE patients associates with clinical efficacy and safety. Further data are forthcoming from ongoing phase III clinical trials of anifrolumab. Other drug development efforts should be considered that target plasmacytoid dendritic cells and toll like receptors given the effects these components have on interferon production.
Collapse
|
71
|
Abstract
The diversity of clinical presentations of lupus nephritis parallel the diversity of pathologic lesions seen in the kidneys of patients with SLE. Renal manifestations range from asymptomatic hematuria or proteinuria to overt nephritic and nephrotic syndromes, rapidly progressive glomerulonephritis, and chronic renal failure. Subclinical nephropathy both during presentation and during monitoring of disease activity is frequently missed because of the notorious unreliability of routine screening urinalyses performed in high-throughput clinical pathology laboratories. Requisitions for urine microscopy should be flagged for special attention in patients at risk for lupus nephritis. Depression of classic complement pathway components and high titers of anti-DNA, anti-nucleosome, or anti-C1q antibodies identify patients are increased risk of renal involvement or flares of nephritis. Several disease activity and damage indexes are available, but they are mostly used in clinical research setting and none has achieved wide use for standard clinical practice.
Collapse
Affiliation(s)
- J E Balow
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892-1818, USA.
| |
Collapse
|
72
|
Abstract
This brief review is focused on different methodologies available for detection of anti-dsDNA antibodies with respect to the best adequacy between biological results of laboratory and clinical significance. A large array of assays has been developed for the measurement of anti-dsDNA. New assays continually introduced have reflected not only technical innovations to avoid difficulties of some assays, but even more with hope to correlate better with systemic lupus erythematosus (SLE), particularly with its clinical course and exacerbations. Finally, with the development of micro arrays technology new insights into the pathophysiology of autoimmune disease processes should be revealed.
Collapse
Affiliation(s)
- A M Rouquette
- Laboratory of Hematology, Tenon Hospital, Pierre et Marie Curie University, Paris, France.
| | | |
Collapse
|
73
|
Pham BN, Albarede S, Guyard A, Burg E, Maisonneuve P. Impact of external quality assessment on antinuclear antibody detection performance. Lupus 2016; 14:113-9. [PMID: 15751815 DOI: 10.1191/0961203305lu2069oa] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Our objective was to evaluate performance of the clinical laboratories for the detection of antinuclear antibodies (ANA) by using indirect immunofluorescence method (IIF), in France. A national external quality assessment (EQA) on ANA detection was organized by the French health products safety agency once a year since 1998. Between 606 to 687 laboratories together with six university reference laboratories experienced in performing tests in autoimmunity participated in the six-year consecutive survey. Each laboratory had to answer to methodological procedures and give coded responses. Variability in IIF methodological procedure was observed. Use of inappropriate microscope magnifications for reading slides or nonconventional cutoff dilution of serum were pointed out to concerned laboratories. Concerning ANA measurement, the rate of good responses ranged from 92.7% to 99.5% of the laboratories when the samples contained ANA. A wide dispersion of ANA titers obtained on a same sample was repeatly observed every year. Misinterpretation of particular fluorescence pattern was noticed. On ANA negative sample, the rate of good responses was 94.3%. In conclusion, ANA detection in routine practice is far from being standardized. However, EQA may have an impact on ANA detection performance when it is conducted on several consecutive year surveys, by providing advice for participating laboratories to limit inter laboratory variations related to methodological procedures.
Collapse
Affiliation(s)
- B-N Pham
- Service d'Hématologie et Immunologie, Hôpital Beaujon, France.
| | | | | | | | | |
Collapse
|
74
|
Kalsi JK, Grossman J, Kim J, Sieling P, Gjertson DW, Reed EF, Ebling FM, Linker-Israeli M, Hahn BH. Peptides from antibodies to DNA elicit cytokine release from peripheral blood mononuclear cells of patients with systemic lupus erythematosus: relation of cytokine pattern to disease duration. Lupus 2016; 13:490-500. [PMID: 15352419 DOI: 10.1191/0961203303lu1060oa] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Peptides from VH regions of antibodies to DNA drive immune responses in systemic lupus erythematosus (SLE). We studied peptide-induced cytokine release by peripheral blood mononuclear cells (PBMC) of patients, the influence of peptide concentration, disease characteristics and HLA-D haplotypes. Cells secreting cytokines (IFNg, IL-2, IL-4 and IL-10) were measured by ELISPOT in PBMC from 31 patients with SLE and 20 matched healthy controls in response to seven peptides (A-G) from the CDR1/FR2 to CDR2/FR3 VH regions of human anti-DNA MAbs. Disease activity was assessed by SELENA-SLEDAI. HLA-DR and -DQ alleles were determined by molecular typing techniques. PBMC from significantly higher proportions of SLE patients than controls responded to VH peptides by generating IFNg and IL-10. Type of cytokines released in response to at least one peptide (D) depended on antigen concentration. Cytokine release was not associated with clinical features of SLE except for disease duration. A shift occurred from IFNg, IL-4 and IL-10 production in early disease to IL-4 and IL-10 in late disease (suggesting increasing TH2-like responses over time). Three peptides (B, D, G) were more stimulatory in the SLE patients than controls. Although none of the peptides was restricted by any particular MHC class II allele, among responders there was increased prevalence of HLA-DQB1 0201 and/or DRB1 0301, alleles known to predispose to SLE. Thus, responses to some VH peptides are more frequent in SLE and vary with disease duration. Increased responses in individuals with HLA class II genotypes that predispose to SLE suggest that peptide presentation by those molecules permits brisker peripheral blood cell responses to autoantibody peptides, thus increasing risk for disease.
Collapse
Affiliation(s)
- J K Kalsi
- Division of Rheumatology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
López-Hoyos M, Cabeza R, Martínez-Taboada VM, Crespo J, SanSegundo D, Blanco R, López-Escribano H, Peña M, Rodríguez-Valverde V. Clinical disease activity and titers of anti-dsDNA antibodies measured by an automated immunofluorescence assay in patients with systemic lupus erythematosus. Lupus 2016; 14:505-9. [PMID: 16130504 DOI: 10.1191/0961203305lu2130oa] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Autoantibodies specific for double stranded DNA (anti-dsDNA Abs) are a serological biomarker of systemic lupus erythematosus (SLE) and constitute useful tools for monitoring many SLE patients. A new automated immunofluorescence and quantitative assay (EliA dsDNA) has recently become available. Its performance has been demonstrated to be equivalent to the Farr and Crithidia luciliae fluorescence (CLIFT) tests. The aim of the present work was to assess the utility of this new assay to monitor clinical activity in a large cohort of SLE patients. To this end, 1020 sera from 181 SLE patients were evaluated by the two methods. Results showed a higher frequency of positive results of anti-dsDNA Abs during lupus flares measured by EliA dsDNA than by CLIFT. Likewise, titers of those Abs were significantly increased in active SLE in comparison with inactive SLE when measured by EliA dsDNA but not by CLIFT. Serum titers of anti-dsDNA Abs by both assays showed a significant negative association with concentrations of C3 and C4. In summary, this retrospective study on a large cohort of patients demonstrated that EliA dsDNA was at least as useful as CLIFT as monitoring tool in the follow-up of SLE patients, but with the advantages of being automated, quick and quantitative.
Collapse
Affiliation(s)
- M López-Hoyos
- Division of Immunology, Hospital Universitario Marqués de Valdecilla, Facultad de Medicina, Universidad de Cantabria, Santander, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Tamura Y, Yoneda A, Takei N, Sawada K. Spatiotemporal Regulation of Hsp90-Ligand Complex Leads to Immune Activation. Front Immunol 2016; 7:201. [PMID: 27252703 PMCID: PMC4877505 DOI: 10.3389/fimmu.2016.00201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/09/2016] [Indexed: 12/19/2022] Open
Abstract
Although heat shock proteins (HSPs) primarily play a pivotal role in the maintenance of cellular homeostasis while reducing extracellular as well as intracellular stresses, their role in immunologically relevant scenarios, including activation of innate immunity as danger signals, antitumor immunity, and autoimmune diseases, is now gaining much attention. The most prominent feature of HSPs is that they function both in their own and as an HSP–ligand complex. We here show as a unique feature of extracellular HSPs that they target chaperoned molecules into a particular endosomal compartment of dendritic cells, thereby inducing innate and adaptive immune responses via spatiotemporal regulation.
Collapse
Affiliation(s)
- Yasuaki Tamura
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Akihiro Yoneda
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Norio Takei
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Kaori Sawada
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| |
Collapse
|
77
|
Akberova NI, Zhmurov AA, Nevzorova TA, Litvinov RI. Molecular dynamics of immune complex of photoadduct-containing DNA with Fab-Anti-DNA antibody fragment. Mol Biol 2016. [DOI: 10.1134/s0026893316020023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
78
|
Stearns NA, Pisetsky DS. The role of monogamous bivalency and Fc interactions in the binding of anti-DNA antibodies to DNA antigen. Clin Immunol 2016; 166-167:38-47. [PMID: 27083935 DOI: 10.1016/j.clim.2016.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 11/18/2022]
Abstract
Antibodies to DNA (anti-DNA) are the serological hallmark of systemic lupus erythematosus. These antibodies can bind DNA avidly by monogamous bivalency, a mechanism which requires the interaction of both Fab combining regions with antigenic determinants on the same polynucleotide. To explore further this mechanism, we tested Fab and F(ab')2 fragments prepared from IgG from patient plasmas in an ELISA with native DNA antigen, detecting antibody with a peroxidase conjugated anti-Fab reagent. These studies showed that Fab fragments, which can only bind monovalently, had negligible activity. Although bivalent F(ab')2 fragments would be predicted to bind DNA, these fragments also showed poor anti-DNA activity. Control studies showed that the fragments retained antibody activity to tetanus toxoid and an EBV antigen preparation. Together, these findings suggest that anti-DNA avidity depends on monogamous bivalency, with the antibody Fc portion also influencing DNA binding, in a mechanism which can be termed Fc-dependent monogamous bivalency.
Collapse
Affiliation(s)
- Nancy A Stearns
- Division of Rheumatology and Immunology, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Medical Research Service, VA Medical Center, Durham, NC, USA
| | - David S Pisetsky
- Division of Rheumatology and Immunology, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Medical Research Service, VA Medical Center, Durham, NC, USA.
| |
Collapse
|
79
|
Epigenetics and innate immunity: the ‘unTolld’ story. Immunol Cell Biol 2016; 94:631-9. [DOI: 10.1038/icb.2016.24] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
|
80
|
Zhao J, Giles BM, Taylor RL, Yette GA, Lough KM, Ng HL, Abraham LJ, Wu H, Kelly JA, Glenn SB, Adler AJ, Williams AH, Comeau ME, Ziegler JT, Marion M, Alarcón-Riquelme ME, Alarcón GS, Anaya JM, Bae SC, Kim D, Lee HS, Criswell LA, Freedman BI, Gilkeson GS, Guthridge JM, Jacob CO, James JA, Kamen DL, Merrill JT, Sivils KM, Niewold TB, Petri MA, Ramsey-Goldman R, Reveille JD, Scofield RH, Stevens AM, Vilá LM, Vyse TJ, Kaufman KM, Harley JB, Langefeld CD, Gaffney PM, Brown EE, Edberg JC, Kimberly RP, Ulgiati D, Tsao BP, Boackle SA. Preferential association of a functional variant in complement receptor 2 with antibodies to double-stranded DNA. Ann Rheum Dis 2016; 75:242-52. [PMID: 25180293 PMCID: PMC4717392 DOI: 10.1136/annrheumdis-2014-205584] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/30/2014] [Accepted: 08/02/2014] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE; OMIM 152700) is characterised by the production of antibodies to nuclear antigens. We previously identified variants in complement receptor 2 (CR2/CD21) that were associated with decreased risk of SLE. This study aimed to identify the causal variant for this association. METHODS Genotyped and imputed genetic variants spanning CR2 were assessed for association with SLE in 15 750 case-control subjects from four ancestral groups. Allele-specific functional effects of associated variants were determined using quantitative real-time PCR, quantitative flow cytometry, electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP)-PCR. RESULTS The strongest association signal was detected at rs1876453 in intron 1 of CR2 (pmeta=4.2×10(-4), OR 0.85), specifically when subjects were stratified based on the presence of dsDNA autoantibodies (case-control pmeta=7.6×10(-7), OR 0.71; case-only pmeta=1.9×10(-4), OR 0.75). Although allele-specific effects on B cell CR2 mRNA or protein levels were not identified, levels of complement receptor 1 (CR1/CD35) mRNA and protein were significantly higher on B cells of subjects harbouring the minor allele (p=0.0248 and p=0.0006, respectively). The minor allele altered the formation of several DNA protein complexes by EMSA, including one containing CCCTC-binding factor (CTCF), an effect that was confirmed by ChIP-PCR. CONCLUSIONS These data suggest that rs1876453 in CR2 has long-range effects on gene regulation that decrease susceptibility to lupus. Since the minor allele at rs1876453 is preferentially associated with reduced risk of the highly specific dsDNA autoantibodies that are present in preclinical, active and severe lupus, understanding its mechanisms will have important therapeutic implications.
Collapse
Affiliation(s)
- Jian Zhao
- Division of Rheumatology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Brendan M Giles
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rhonda L Taylor
- School of Pathology and Laboratory Medicine, Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia
| | - Gabriel A Yette
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kara M Lough
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Han Leng Ng
- School of Pathology and Laboratory Medicine, Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia
| | - Lawrence J Abraham
- School of Pathology and Laboratory Medicine, Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia
| | - Hui Wu
- Division of Rheumatology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Jennifer A Kelly
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Stuart B Glenn
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Adam J Adler
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Adrienne H Williams
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mary E Comeau
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Julie T Ziegler
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Miranda Marion
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Marta E Alarcón-Riquelme
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Pfizer-Universidad de Granada-Junta de Andalucía Center for Genomics and Oncological Research, Granada, Spain
| | | | - Graciela S Alarcón
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), Universidad del Rosario, Bogotá, Colombia
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Dam Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Lindsey A Criswell
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California San Francisco, San Francisco, California, USA
| | - Barry I Freedman
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Gary S Gilkeson
- Division of Rheumatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joel M Guthridge
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Chaim O Jacob
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | - Judith A James
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Diane L Kamen
- Division of Rheumatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joan T Merrill
- Department of Clinical Pharmacology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kathy Moser Sivils
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Timothy B Niewold
- Division of Rheumatology and Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michelle A Petri
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rosalind Ramsey-Goldman
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - John D Reveille
- Department of Rheumatology and Clinical Immunogenetics, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - R Hal Scofield
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- US Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| | - Anne M Stevens
- Division of Rheumatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Luis M Vilá
- Division of Rheumatology, Department of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Timothy J Vyse
- Division of Genetics and Molecular Medicine and Immunology, King's College London, London, UK
| | - Kenneth M Kaufman
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - John B Harley
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Carl D Langefeld
- Department of Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Elizabeth E Brown
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeffrey C Edberg
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert P Kimberly
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daniela Ulgiati
- School of Pathology and Laboratory Medicine, Centre for Genetic Origins of Health and Disease, The University of Western Australia, Crawley, Western Australia, Australia
| | - Betty P Tsao
- Division of Rheumatology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Susan A Boackle
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Denver Veterans Affairs Medical Center, Denver, Colorado, USA
| |
Collapse
|
81
|
Wang J, Lon HK, Lee SL, Burckart GJ, Pisetsky DS. Oligonucleotide-Based Drug Development: Considerations for Clinical Pharmacology and Immunogenicity. Ther Innov Regul Sci 2015; 49:861-868. [PMID: 30222372 DOI: 10.1177/2168479015592195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The field of oligonucleotide (OGN)-based therapeutics has been growing dramatically in the past decade, providing innovative platforms to develop agents for the treatment of a wide variety of clinical conditions. OGN agents have unique physicochemical properties and pharmacokinetic/pharmacodynamic characteristics. This review considers findings from the literature and information on new molecular entities submitted to the US Food and Drug Administration as OGN-based therapeutics. In addition, the article discusses several challenging issues from the perspective of clinical pharmacology, emphasizing the potential of immunogenicity, the effect of renal impairment on OGN exposure, drug-drug interactions, and the utility of pharmacokinetic/pharmacodynamic modeling. The field of OGN-based therapeutics is in evolution and will benefit from further studies as well as clinical experience to formulate guidelines and promote the development of this class of agents.
Collapse
Affiliation(s)
- Jian Wang
- 1 Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, MD, USA
| | - Hoi-Kei Lon
- 1 Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, MD, USA.,2 Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Shwu-Luan Lee
- 3 Office of Hematology and Oncology Products, Office of New Drugs, US Food and Drug Administration, Silver Spring, MD, USA
| | - Gilbert J Burckart
- 1 Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, MD, USA
| | - David S Pisetsky
- 4 Medical Research Service, Durham VA Medical Center and Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
82
|
Sánchez B, Hevia A, González S, Margolles A. Interaction of Intestinal Microorganisms with the Human Host in the Framework of Autoimmune Diseases. Front Immunol 2015; 6:594. [PMID: 26635808 PMCID: PMC4653298 DOI: 10.3389/fimmu.2015.00594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/05/2015] [Indexed: 01/03/2023] Open
Abstract
Autoimmune diseases, such as systemic lupus erythematosus (SLE), are caused by a complex interaction of environmental-, genetic-, and sex-related factors. Although SLE has traditionally been considered independent from the microbiota, recent work published during the last 5 years suggests a strong connection between SLE and the composition of our gut commensals as one of the main environmental factors linked to this disease. Preliminary data have evidenced that (i) interaction of certain microbial-derived molecules with specific cell receptors and (ii) the influence of certain commensal microorganisms over specific immune cell subsets plays an important role in the pathogenesis of SLE and SLE-like diseases. In addition, epigenetic changes driven by certain microbial groups have been recently proposed as an additional link between gut microbiota and SLE. As immune responses elicited against commensal bacteria are deeply dependent on the composition of the latter, and as microbial populations can be modified by dietary interventions, identifying the precise gut microorganisms responsible for worsening the SLE symptoms is of crucial importance for this and other SLE-related diseases, including antiphospholipid syndrome or lupus nephritis. In this minireview, the current knowledge on the relationships between microbes and SLE and SLE-related diseases is compiled and discussed.
Collapse
Affiliation(s)
- Borja Sánchez
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| | - Arancha Hevia
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| | - Sonia González
- Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| |
Collapse
|
83
|
Abstract
Antibodies that recognize and bind to DNA (anti-DNA antibodies) are serological hallmarks of systemic lupus erythematosus (SLE) and key markers for diagnosis and disease activity. In addition to common use in the clinic, anti-DNA antibody testing now also determines eligibility for clinical trials, raising important questions about the nature of the antibody-antigen interaction. At present, no 'gold standard' for serological assessment exists, and anti-DNA antibody binding can be measured with a variety of assay formats, which differ in the nature of the DNA substrates and in the conditions for binding and detection of antibodies. A mechanism called monogamous bivalency--in which high avidity results from simultaneous interaction of IgG Fab sites with a single polynucleotide chain--determines anti-DNA antibody binding; this mechanism might affect antibody detection in different assay formats. Although anti-DNA antibodies can promote pathogenesis by depositing in the kidney or driving cytokine production, they are not all alike, pathologically, and anti-DNA antibody expression does not necessarily correlate with active disease. Levels of anti-DNA antibodies in patients with SLE can vary over time, distinguishing anti-DNA antibodies from other pathogenic antinuclear antibodies. Elucidation of the binding specificities and the pathogenic roles of anti-DNA antibodies in SLE should enable improvements in the design of informative assays for both clinical and research purposes.
Collapse
Affiliation(s)
- David S Pisetsky
- Medical Research Service, Durham Veterans Administration Medical Center, Box 151G, 508 Fulton Street, Durham, North Carolina 27705, USA
| |
Collapse
|
84
|
Antibodies as Mediators of Brain Pathology. Trends Immunol 2015; 36:709-724. [PMID: 26494046 DOI: 10.1016/j.it.2015.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/17/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The brain is normally sequestered from antibody exposure by the blood brain barrier. However, antibodies can access the brain during fetal development before the barrier achieves full integrity, and in disease states when barrier integrity is compromised. Recent studies suggest that antibodies contribute to brain pathology associated with autoimmune diseases such as systemic lupus erythematosus and neuromyelitis optica, and can lead to transient or permanent behavioral or cognitive abnormalities. We review these findings here and examine the circumstances associated with antibody entry into the brain, the routes of access and the mechanisms that then effect pathology. Understanding these processes and the nature and specificity of neuronal autoantibodies may reveal therapeutic strategies toward alleviating or preventing the neurological pathologies and behavioral abnormalities associated with autoimmune disease.
Collapse
|
85
|
Iruretagoyena M, Hirigoyen D, Naves R, Burgos PI. Immune Response Modulation by Vitamin D: Role in Systemic Lupus Erythematosus. Front Immunol 2015; 6:513. [PMID: 26528285 PMCID: PMC4600954 DOI: 10.3389/fimmu.2015.00513] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
Vitamin D plays key roles as a natural immune modulator and has been implicated in the pathophysiology of autoimmune diseases, including systemic lupus erythematosus (SLE). This review presents a summary and analysis of the recent literature regarding immunoregulatory effects of vitamin D as well as its importance in SLE development, clinical severity, and possible effects of supplementation in disease treatment.
Collapse
Affiliation(s)
- Mirentxu Iruretagoyena
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Daniela Hirigoyen
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Rodrigo Naves
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile , Santiago , Chile
| | - Paula Isabel Burgos
- Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
86
|
Im SR, Im SW, Chung HY, Pravinsagar P, Jang YJ. Cell- and nuclear-penetrating anti-dsDNA autoantibodies have multiple arginines in CDR3 of VH and increase cellular level of pERK and Bcl-2 in mesangial cells. Mol Immunol 2015; 67:377-87. [DOI: 10.1016/j.molimm.2015.06.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 01/27/2023]
|
87
|
Yung S, Chan TM. Mechanisms of Kidney Injury in Lupus Nephritis - the Role of Anti-dsDNA Antibodies. Front Immunol 2015; 6:475. [PMID: 26441980 PMCID: PMC4569852 DOI: 10.3389/fimmu.2015.00475] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/01/2015] [Indexed: 01/12/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by a breakdown of self-tolerance, production of auto-antibodies and immune-mediated injury, resulting in damage accrual in multiple organs. Kidney involvement, termed lupus nephritis, is a major cause of morbidity and mortality that affects over half of the SLE population during the course of disease. The etiology of lupus nephritis is multifactorial and remains to be fully elucidated. Accumulating evidence suggests that in addition to forming immune complexes and triggering complement activation, anti-dsDNA antibodies contribute to the pathogenesis of lupus nephritis through binding, either directly or indirectly, to cross-reactive antigens or chromatin materials, respectively, to resident renal cells and/or extracellular matrix components, thereby triggering downstream cellular activation and proliferation as well as inflammatory and fibrotic processes. Several cross-reactive antigens that mediate anti-dsDNA antibody binding have been identified, such as annexin II and alpha-actinin. This review discusses the mechanisms through which anti-dsDNA antibodies contribute to immunopathogenesis in lupus nephritis. Corticosteroids combined with either mycophenolic acid (MPA) or cyclophosphamide is the current standard of care immunosuppressive therapy for severe lupus nephritis. This review also discusses recent data showing distinct effects of MPA and cyclophosphamide on inflammatory and fibrotic processes in resident renal cells.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong , Hong Kong , China
| | - Tak Mao Chan
- Department of Medicine, Queen Mary Hospital, University of Hong Kong , Hong Kong , China
| |
Collapse
|
88
|
Sex differences in the early life correlates of natural antibody concentrations. J Dev Orig Health Dis 2015; 6:501-11. [PMID: 26279187 DOI: 10.1017/s2040174415001373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Innate-like B1a lymphocytes arise from long-lived progenitors produced exclusively by fetal stem cells. Any insults coinciding with this early lymphopoietic wave could have a permanent impact on the B1a population and its unique protein products, the natural antibodies (NAb). We investigated early life nutritional influences on NAb concentrations of pre-adolescent children (n=290) in rural Nepal for whom we had extensive information on exposures from pregnancy and early infancy. Infant size and growth were strongly associated with NAb concentrations at 9-13 years of age among males (e.g., for neonatal weight: βBOYS=0.43; P<0.001), but not females (e.g., for neonatal weight: βGIRLS=-0.16; P=0.26). In females, season of birth was associated with NAb concentrations, with marked reductions among girls born during the pre-monsoon (March-May; βGIRLS=-0.39; P=0.01) and pre-harvest (September-November; βGIRLS=-0.35; P=0.03) seasons. Our findings suggest that nutritional or other environmental influences on immune development may vary by sex, with potential consequences for immune function during infancy and long-term risk of immune-mediated disease.
Collapse
|
89
|
Williams JM, Bonami RH, Hulbert C, Thomas JW. Reversing Tolerance in Isotype Switch-Competent Anti-Insulin B Lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:853-64. [PMID: 26109644 PMCID: PMC4506889 DOI: 10.4049/jimmunol.1403114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 05/30/2015] [Indexed: 12/22/2022]
Abstract
Autoreactive B lymphocytes that escape central tolerance and mature in the periphery are a liability for developing autoimmunity. IgG insulin autoantibodies that predict type 1 diabetes and complicate insulin therapies indicate that mechanisms for tolerance to insulin are flawed. To examine peripheral tolerance in anti-insulin B cells, we generated C57BL/6 mice that harbor anti-insulin VDJH-125 site directed to the native IgH locus (VH125(SD)). Class switch-competent anti-insulin B cells fail to produce IgG Abs following T cell-dependent immunization of VH125(SD) mice with heterologous insulin, and they exhibit markedly impaired proliferation to anti-CD40 plus insulin in vitro. In contrast, costimulation with LPS plus insulin drives robust anti-insulin B cell proliferation. Furthermore, VH125(SD) mice produce both IgM and IgG2a anti-insulin Abs following immunization with insulin conjugated to type 1 T cell-independent Brucella abortus ring test Ag (BRT). Anti-insulin B cells undergo clonal expansion in vivo and emerge as IgM(+) and IgM(-) GL7(+)Fas(+) germinal center (GC) B cells following immunization with insulin-BRT, but not BRT alone. Analysis of Igκ genes in VH125(SD) mice immunized with insulin-BRT reveals that anti-insulin Vκ from the preimmune repertoire is selected into GCs. These data demonstrate that class switch-competent anti-insulin B cells remain functionally silent in T cell-dependent immune responses, yet these B cells are vulnerable to reversal of anergy following combined BCR/TLR engagement that promotes Ag-specific GC responses and Ab production. Environmental factors that lead to infection and inflammation could play a critical yet underappreciated role in driving loss of tolerance and promoting autoimmune disease.
Collapse
Affiliation(s)
- Jonathan M Williams
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Chrys Hulbert
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| | - James W Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; and Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
90
|
Thiyagarajan D, Rekvig OP, Seredkina N. TNFα Amplifies DNaseI Expression in Renal Tubular Cells while IL-1β Promotes Nuclear DNaseI Translocation in an Endonuclease-Inactive Form. PLoS One 2015; 10:e0129485. [PMID: 26065428 PMCID: PMC4465975 DOI: 10.1371/journal.pone.0129485] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 05/08/2015] [Indexed: 11/18/2022] Open
Abstract
We have demonstrated that the renal endonuclease DNaseI is up-regulated in mesangial nephritis while down-regulated during progression of the disease. To determine the basis for these reciprocal DNaseI expression profiles we analyse processes accounting for an early increase in renal DNaseI expression. Main hypotheses were that i. the mesangial inflammation and secreted pro-inflammatory cytokines directly increase DNaseI protein expression in tubular cells, ii. the anti-apoptotic protein tumor necrosis factor receptor-associated protein 1 (Trap 1) is down-regulated by increased expression of DNaseI due to transcriptional interference, and iii. pro-inflammatory cytokines promote nuclear translocation of a variant of DNaseI. The latter hypothesis emerges from the fact that anti-DNaseI antibodies stained tubular cell nuclei in murine and human lupus nephritis. The present study was performed on human tubular epithelial cells stimulated with pro-inflammatory cytokines. Expression of the DNaseI and Trap 1 genes was determined by qPCR, confocal microscopy, gel zymography, western blot and by immune electron microscopy. Results from in vitro cell culture experiments were analysed for biological relevance in kidneys from (NZBxNZW)F1 mice and human patients with lupus nephritis. Central data indicate that stimulating the tubular cells with TNFα promoted increased DNaseI and reduced Trap 1 expression, while TNFα and IL-1β stimulation induced nuclear translocation of the DNaseI. TNFα-stimulation resulted in 3 distinct effects; increased DNaseI and IL-1β gene expression, and nuclear translocation of DNaseI. IL-1β-stimulation solely induced nuclear DNaseI translocation. Tubular cells stimulated with TNFα and simultaneously transfected with IL-1β siRNA resulted in increased DNaseI expression but no nuclear translocation. This demonstrates that IL-1β promotes nuclear translocation of a cytoplasmic variant of DNaseI since translocation clearly was not dependent on DNaseI gene activation. Nuclear translocated DNaseI is shown to be enzymatically inactive, which may point at a new, yet unknown function of renal DNaseI.
Collapse
Affiliation(s)
- Dhivya Thiyagarajan
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ole Petter Rekvig
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Natalya Seredkina
- RNA and Molecular Pathology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
91
|
He C, Mao T, Feng Y, Song T, Qin C, Yan R, Feng P. Anti-CII antibody as a novel indicator to assess disease activity in systemic lupus erythematosus. Lupus 2015; 24:1370-6. [PMID: 26048288 DOI: 10.1177/0961203315588970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 05/06/2015] [Indexed: 11/16/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that affects a variety of organ systems. Anti-dsDNA Abs and complement factors have been used as indicators of lupus activity for more than 50 years. A novel indicator of activation in SLE is reported in this paper. Anti-collagen type II (CII) Ab was obviously elevated in patients with SLE compared to those patients with ankylosing spondylitis (AS) and healthy controls (HCs). Anti-CII-Ab-positive patients with SLE showed significantly higher levels of serum IgG and higher titers of ANA but lower levels of C3 and C4 than controls. A positive correlation was demonstrated between anti-CII Ab and serum IgG in SLE patients (r = 0.50, p < 0.0001). The negative correlations of anti-CII Ab with C3 and C4 were observed in SLE patients (r = -0.36, p = 0.0013; r = -0.37, p = 0.0006, respectively). The reduced anti-CII Ab level was accompanied by decreased level of serum IgG and increased levels of C3 and C4 after regular treatment. Therefore, anti-CII Ab could be a novel indicator for monitoring activity of SLE.
Collapse
Affiliation(s)
- C He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - T Mao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Y Feng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - T Song
- Department of Clinical Nutrition, 302 Hospital of PLA, Beijing, China
| | - C Qin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - R Yan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - P Feng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
92
|
Abstract
The inclusion of 'the anti-DNA antibody' by the ACR and the Systemic Lupus International Collaborating Clinics (SLICC) as a criterion for systemic lupus erythematosus does not convey the diverse origins of these antibodies, whether their production is transient or persistent (which is heavily influenced by the nature of the inducing antigens), the specificities exerted by these antibodies or their clinical impact-or lack thereof. A substantial amount of data not considered in clinical medicine could be added from basic immunology evidence, which could change the paradigms linked to what 'the anti-DNA antibody' is, in a pathogenic, classification or diagnostic context.
Collapse
|
93
|
HMGB1 Promotes Systemic Lupus Erythematosus by Enhancing Macrophage Inflammatory Response. J Immunol Res 2015; 2015:946748. [PMID: 26078984 PMCID: PMC4452473 DOI: 10.1155/2015/946748] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/28/2014] [Indexed: 01/06/2023] Open
Abstract
Background/Purpose. HMGB1, which may act as a proinflammatory mediator, has been proposed to contribute to the pathogenesis of multiple chronic inflammatory and autoimmune diseases including systemic lupus erythematosus (SLE); however, the precise mechanism of HMGB1 in the pathogenic process of SLE remains obscure. Method. The expression of HMGB1 was measured by ELISA and western blot. The ELISA was also applied to detect proinflammatory cytokines levels. Furthermore, nephritic pathology was evaluated by H&E staining of renal tissues. Results. In this study, we found that HMGB1 levels were significantly increased and correlated with SLE disease activity in both clinical patients and murine model. Furthermore, gain- and loss-of-function analysis showed that HMGB1 exacerbated the severity of SLE. Of note, the HMGB1 levels were found to be associated with the levels of proinflammatory cytokines such as TNF-α and IL-6 in SLE patients. Further study demonstrated that increased HMGB1 expression deteriorated the severity of SLE via enhancing macrophage inflammatory response. Moreover, we found that receptor of advanced glycation end products played a critical role in HMGB1-mediated macrophage inflammatory response. Conclusion. These findings suggested that HMGB1 might be a risk factor for SLE, and manipulation of HMGB1 signaling might provide a therapeutic strategy for SLE.
Collapse
|
94
|
Systemic Lupus Erythematosus with and without Anti-dsDNA Antibodies: Analysis from a Large Monocentric Cohort. Mediators Inflamm 2015; 2015:328078. [PMID: 26063969 PMCID: PMC4438145 DOI: 10.1155/2015/328078] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/02/2015] [Indexed: 12/05/2022] Open
Abstract
Objectives. The anti-dsDNA antibodies are a marker for Systemic Lupus Erythematosus (SLE) and 70–98% of patients test positive. We evaluated the demographic, clinical, laboratory, and therapeutical features of a monocentric SLE cohort according to the anti-dsDNA status. Methods. We identified three groups: anti-dsDNA + (persistent positivity); anti-dsDNA ± (initial positivity and subsequent negativity during disease course); anti-dsDNA − (persistent negativity). Disease activity was assessed by the European Consensus Lupus Activity Measurement (ECLAM). Results. We evaluated 393 patients (anti-dsDNA +: 62.3%; anti-dsDNA ±: 13.3%; anti-dsDNA −: 24.4%). The renal involvement was significantly more frequent in anti-dsDNA + (30.2%), compared with anti-dsDNA ± and anti-dsDNA − (21.1% and 18.7%, resp.; P = 0.001). Serositis resulted significantly more frequent in anti-dsDNA − (82.3%) compared to anti-dsDNA + and anti-dsDNA ± (20.8% and 13.4%, resp.; P < 0.0001). The reduction of C4 serum levels was identified significantly more frequently in anti-dsDNA + and anti-dsDNA ± (40.0% and 44.2%, resp.) compared with anti-dsDNA − (21.8%, P = 0.005). We did not identify significant differences in the mean ECLAM values before and after modification of anti-dsDNA status (P = 0.7). Conclusion. Anti-dsDNA status influences the clinical and immunological features of SLE patients. Nonetheless, it does not appear to affect disease activity.
Collapse
|
95
|
Iqbal M, Bashir S, Al-Ayadhi L. Prevalence of antimitochondrial antibodies in autism spectrum subjects. FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ABSTRACT Objective: Autism is a neurodevelopmental disorder characterized by impairment in verbal and nonverbal communication, repetitive and stereotypic behavior. Dysregulated immune system has a role in the pathogenesis of Autism. This study was designed to measure the prevalence of antimitochondrial (AM) antibodies in a group of autistic children. Methods: AM antibodies subtype 2 (AMA-M2) were evaluated by indirect solid phase enzyme immunoassay in 62 autistic children and 14 age-matched healthy controls. Autistic activity was assessed by using the Childhood Autism Rating Scale. Results: Significantly elevated levels of AMA-M2 were observed in the sera of autistic children (n = 54, 0.221 ± 0.029 IU/ml [mean ± SEM]) compared with healthy controls (n = 14, 0.111 ± 0.010 IU/ml [mean ± SEM], p = 0.0008) and there was no significant difference in patients with moderate to severe autism (p = 0.49). AM antibodies in autistic patients have no correlation with Childhood Autism Rating Scale score. Conclusion: The current study demonstrated significantly high levels of AMA-M2 in autistic subjects when compared with healthy controls. Further large-scale studies are required to dissect any pathogenic role of these antibodies in the development of autism.
Collapse
Affiliation(s)
- Muhammad Iqbal
- Aging Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, PO box 2925, Riyadh 11461, Saudi Arabia
| | - Shahid Bashir
- KSU-Autism Research & Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, PO box 2925, Riyadh 11461, Saudi Arabia
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laila Al-Ayadhi
- KSU-Autism Research & Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, Faculty of Medicine, King Saud University, PO box 2925, Riyadh 11461, Saudi Arabia
| |
Collapse
|
96
|
Saito K, Kukita K, Kutomi G, Okuya K, Asanuma H, Tabeya T, Naishiro Y, Yamamoto M, Takahashi H, Torigoe T, Nakai A, Shinomura Y, Hirata K, Sato N, Tamura Y. Heat shock protein 90 associates with Toll-like receptors 7/9 and mediates self-nucleic acid recognition in SLE. Eur J Immunol 2015; 45:2028-41. [PMID: 25871979 DOI: 10.1002/eji.201445293] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/26/2015] [Accepted: 04/09/2015] [Indexed: 01/09/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototype systemic autoimmune disease, and disease activity is associated with serum IFN-α level. Plasmacytoid dendritic cells (pDCs) sense microbial as well as self-nucleic acids by TLRs 7 and 9 and produce a large amount of IFN-α. Here, we show that heat shock protein 90 (Hsp90) associates with and delivers TLR7/9 from the ER to early endosomes for ligand recognition. Inhibition of Hsp90 by various approaches including the use of Hsp90 inhibitor, a geldanamycin derivative, suppressed the Hsp90 association with TLR7/9, which resulted in inhibition of IFN-α production, leading to improvement of SLE symptoms in mice. Notably, we observed that serum Hsp90 is clearly increased in patients with active SLE compared with that in patients with inactive disease. Furthermore, we demonstrated that serum Hsp90 detected in SLE patients binds to self-DNA and/or anti-DNA Ab, thus leading to stimulation of pDCs to produce IFN-α. Our data demonstrate that Hsp90 plays a crucial role in the pathogenesis of SLE and that an Hsp90 inhibitor will therefore provide a new therapeutic approach to SLE and other nucleic acid-related autoimmune diseases.
Collapse
Affiliation(s)
- Keita Saito
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuharu Kukita
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Goro Kutomi
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koichi Okuya
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroko Asanuma
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuya Tabeya
- Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuka Naishiro
- Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Motohisa Yamamoto
- Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Yamaguchi, Japan
| | - Yasuhisa Shinomura
- Department of Internal Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koichi Hirata
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuaki Tamura
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Molecular Therapeutics, Center for Food and Medical Innovation, Hokkaido University, Sapporo, Japan
| |
Collapse
|
97
|
Hoyer BF, Dörner T. [Biologics therapy for systemic lupus erythematosus. Current situation]. Z Rheumatol 2015; 74:206-14. [PMID: 25854155 DOI: 10.1007/s00393-014-1458-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Improved understanding of the immunopathogenesis of systemic lupus erythematosus (SLE) has paved the way for new specific immune interventions for this inflammatory disease similar to those for rheumatoid arthritis and spondylarthritides. METHODS New biologics were developed on this basis or are in the process of clinical development and open up new therapy options for patients. In this context belimumab is of particular importance. As an innovative biologic the monoclonal antibody against the cytokine BAFF/BLyS (belimumab) has been approved for the treatment of serologically active SLE. A number of other biologics against other cytokines are in the clinical development phase and appear to be promising for further improvement of the current therapeutic possibilities in SLE. This article addresses the current aspects of immune interventions with biologics for SLE and the specific challenges of this disease.
Collapse
Affiliation(s)
- B F Hoyer
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Deutschland,
| | | |
Collapse
|
98
|
Al Gadban MM, Alwan MM, Smith KJ, Hammad SM. Accelerated vascular disease in systemic lupus erythematosus: role of macrophage. Clin Immunol 2015; 157:133-44. [PMID: 25638414 PMCID: PMC4410070 DOI: 10.1016/j.clim.2015.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory condition that is considered a major cause of death worldwide. Striking phenomena of atherosclerosis associated with systemic lupus erythematosus (SLE) is its high incidence in young patients. Macrophages are heterogeneous cells that differentiate from hematopoietic progenitors and reside in different tissues to preserve tissue integrity. Macrophages scavenge modified lipids and play a major role in the development of atherosclerosis. When activated, macrophages secret inflammatory cytokines. This activation triggers apoptosis of cells in the vicinity of macrophages. As such, macrophages play a significant role in tissue remodeling including atherosclerotic plaque formation and rupture. In spite of studies carried on identifying the role of macrophages in atherosclerosis, this role has not been studied thoroughly in SLE-associated atherosclerosis. In this review, we address factors released by macrophages as well as extrinsic factors that may control macrophage behavior and their effect on accelerated development of atherosclerosis in SLE.
Collapse
Affiliation(s)
- Mohammed M Al Gadban
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Mohamed M Alwan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Kent J Smith
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Samar M Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
99
|
Taddeo A, Khodadadi L, Voigt C, Mumtaz IM, Cheng Q, Moser K, Alexander T, Manz RA, Radbruch A, Hiepe F, Hoyer BF. Long-lived plasma cells are early and constantly generated in New Zealand Black/New Zealand White F1 mice and their therapeutic depletion requires a combined targeting of autoreactive plasma cells and their precursors. Arthritis Res Ther 2015; 17:39. [PMID: 25889236 PMCID: PMC4411657 DOI: 10.1186/s13075-015-0551-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/06/2015] [Indexed: 12/16/2022] Open
Abstract
Introduction Autoantibodies contribute significantly to the pathogenesis of systemic lupus erythematosus (SLE). Unfortunately, the long-lived plasma cells (LLPCs) secreting such autoantibodies are refractory to conventional immunosuppressive treatments. Although generated long before the disease becomes clinically apparent, it remains rather unclear whether LLPC generation continues in the established disease. Here, we analyzed the generation of LLPCs, including autoreactive LLPCs, in SLE-prone New Zealand Black/New Zealand White F1 (NZB/W F1) mice over their lifetime, and their regeneration after depletion. Methods Bromodeoxyuridine pulse-chase experiments in mice of different ages were performed in order to analyze the generation of LLPCs during the development of SLE. LLPCs were enumerated by flow cytometry and autoreactive anti-double-stranded DNA (anti-dsDNA) plasma cells by enzyme-linked immunospot (ELISPOT). For analyzing the regeneration of LLPCs after depletion, mice were treated with bortezomib alone or in combination with cyclophosphamide and plasma cells were enumerated 12 hours, 3, 7, 11 and 15 days after the end of the bortezomib cycle. Results Autoreactive LLPCs are established in the spleen and bone marrow of SLE-prone mice very early in ontogeny, before week 4 and before the onset of symptoms. The generation of LLPCs then continues throughout life. LLPC counts in the spleen plateau by week 10, but continue to increase in the bone marrow and inflamed kidney. When LLPCs are depleted by the proteasome inhibitor bortezomib, their numbers regenerate within two weeks. Persistent depletion of LLPCs was achieved only by combining a cycle of bortezomib with maintenance therapy, for example cyclophosphamide, depleting the precursors of LLPCs or preventing their differentiation into LLPCs. Conclusions In SLE-prone NZB/W F1 mice, autoreactive LLPCs are generated throughout life. Their sustained therapeutic elimination requires both the depletion of LLPCs and the inhibition of their regeneration.
Collapse
Affiliation(s)
- Adriano Taddeo
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Laleh Khodadadi
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Caroline Voigt
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Imtiaz M Mumtaz
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Qingyu Cheng
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Katrin Moser
- German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Ratzeburger Allee 160, 23562, Luebeck, Germany.
| | - Andreas Radbruch
- German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Falk Hiepe
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| | - Bimba F Hoyer
- Department of Rheumatology and Clinical Immunology, Charité University Hospital Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,German Rheumatism Research Centre, a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
100
|
Steiman AJ, Urowitz MB, Ibañez D, Li TT, Gladman DD, Wither J. Anti-dsDNA and Antichromatin Antibody Isotypes in Serologically Active Clinically Quiescent Systemic Lupus Erythematosus. J Rheumatol 2015; 42:810-6. [PMID: 25729033 DOI: 10.3899/jrheum.140796] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Serologically active clinically quiescent (SACQ) patients with systemic lupus erythematosus (SLE) are clinically quiescent despite serologic activity. Since studies suggest that antichromatin antibodies are more sensitive than anti-dsDNA antibodies in detecting active SLE, and that immunoglobulin (Ig) G, in particular complement-fixing subclasses, may be more pathogenic than IgM, we investigated the levels of anti-dsDNA and antichromatin isotypes in SACQ patients as compared to non-SACQ patients with SLE. METHODS Levels of IgM, IgA, IgG, and IgG1-4 antichromatin and anti-dsDNA were measured by ELISA. SACQ was defined as ≥ 2 years with the SLE Disease Activity Index 2000 (SLEDAI-2K) at 2 or 4 from serologic activity, during which patients could be taking antimalarials, but not corticosteroids or immunosuppressives. Unselected non-SACQ patients with SLE were used as comparators. SACQ patient serum samples were further stratified based on subsequent development of flare, defined as clinical SLEDAI-2K ≥ 1 and/or treatment initiation. Nonparametric statistics were used, and generalized estimating equations were applied to account for multiple samples in the same patient. RESULTS SACQ patients' complement-fixing antichromatin and anti-dsDNA IgG subclasses were significantly higher than those of non-SACQ patients. When the sample drawn latest in a SACQ period was analyzed, there was no difference between antichromatin or anti-dsDNA isotype or IgG subclass levels between patients who flared and those who remained SACQ, nor were consistent trends seen when samples were examined during SACQ and flare in the same patient. CONCLUSION The SACQ phenotype does not arise from a lack of pathogenic anti-dsDNA and/or antichromatin autoantibodies. Neither increases in antichromatin nor anti-dsDNA isotype or IgG subclass levels were predictive of or coincident with flare in SACQ patients.
Collapse
Affiliation(s)
- Amanda J Steiman
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Murray B Urowitz
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Dominique Ibañez
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Timothy T Li
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Dafna D Gladman
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network
| | - Joan Wither
- From the University of Toronto; Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital; Toronto Western Hospital; Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.A.J. Steiman, MD, FRCPC, Rheumatology Fellow, University of Toronto, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; M.B. Urowitz, MD, FRCPC, Professor of Medicine, University of Toronto, and Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; D. Ibañez, MSc, Biostatistician, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; T.T. Li, MSc, Medical Student, Arthritis Centre of Excellence, Division of Genetics and Development, Western Hospital Research Institute, University Health Network; D.D. Gladman, MD, FRCPC, Professor of Medicine, University of Toronto, and Deputy Director, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital; J. Wither, MD, PhD, FRCPC, Professor of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network.
| |
Collapse
|