1
|
Azizan A, Farhadi E, Faezi ST, Jamshidi A, Alikhani M, Mahmoudi M. Role of miRNAs in Apoptosis Pathways of Immune Cells in Systemic Lupus Erythematosus. Immun Inflamm Dis 2025; 13:e70124. [PMID: 39912562 PMCID: PMC11800236 DOI: 10.1002/iid3.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by dysregulated immune responses and multi-organ involvement. Dysregulation of apoptosis, a key process for maintaining immune homeostasis, plays a critical role in the pathogenesis of SLE. MicroRNAs (miRNAs), small non-coding RNAs that regulate gene expression, have emerged as important modulators of apoptosis in immune cells, influencing the balance between immune tolerance and autoimmunity. OBJECTIVES This review aims to comprehensively summarize recent advancements in understanding the roles of miRNAs in apoptosis regulation within immune cells in SLE, highlighting their therapeutic potential for restoring immune balance and mitigating disease progression. RESULTS Aberrant expression of specific miRNAs contributes to the dysregulation of apoptosis in SLE immune cells. Pro-apoptotic miRNAs, such as miR-125b and miR-150, are often downregulated, leading to enhanced survival of autoreactive immune cells. Conversely, anti-apoptotic miRNAs, including miR-21, are upregulated, further disrupting the delicate balance of immune cell apoptosis. Dual-function miRNAs, such as miR-155, exhibit context-dependent roles based on cellular environments and target gene interactions. This dysregulation promotes the persistence of autoreactive immune cells and the development of autoimmunity. CONCLUSIONS miRNAs play critical roles in modulating apoptosis pathways, making them promising therapeutic targets for SLE. Restoring the balance of pro-apoptotic and anti-apoptotic miRNAs could help reinstate immune tolerance and reduce tissue damage. Future research should focus on elucidating miRNA targetomes, improving delivery systems, and addressing off-target effects to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Amin Azizan
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Elham Farhadi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | | | - Ahmadreza Jamshidi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Majid Alikhani
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Mahdi Mahmoudi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| |
Collapse
|
2
|
von Hofsten S, Fenton KA, Pedersen HL. Human and Murine Toll-like Receptor-Driven Disease in Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:5351. [PMID: 38791389 PMCID: PMC11120885 DOI: 10.3390/ijms25105351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is linked to the differential roles of toll-like receptors (TLRs), particularly TLR7, TLR8, and TLR9. TLR7 overexpression or gene duplication, as seen with the Y-linked autoimmune accelerator (Yaa) locus or TLR7 agonist imiquimod, correlates with increased SLE severity, and specific TLR7 polymorphisms and gain-of-function variants are associated with enhanced SLE susceptibility and severity. In addition, the X-chromosome location of TLR7 and its escape from X-chromosome inactivation provide a genetic basis for female predominance in SLE. The absence of TLR8 and TLR9 have been shown to exacerbate the detrimental effects of TLR7, leading to upregulated TLR7 activity and increased disease severity in mouse models of SLE. The regulatory functions of TLR8 and TLR9 have been proposed to involve competition for the endosomal trafficking chaperone UNC93B1. However, recent evidence implies more direct, regulatory functions of TLR9 on TLR7 activity. The association between age-associated B cells (ABCs) and autoantibody production positions these cells as potential targets for treatment in SLE, but the lack of specific markers necessitates further research for precise therapeutic intervention. Therapeutically, targeting TLRs is a promising strategy for SLE treatment, with drugs like hydroxychloroquine already in clinical use.
Collapse
Affiliation(s)
- Susannah von Hofsten
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Kristin Andreassen Fenton
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Hege Lynum Pedersen
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| |
Collapse
|
3
|
Soni C, Makita S, Eichinger A, Serpas L, Sisirak V, Reizis B. Cutting Edge: TLR2 Signaling in B Cells Promotes Autoreactivity to DNA via IL-6 Secretion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1475-1480. [PMID: 37800687 PMCID: PMC10841863 DOI: 10.4049/jimmunol.2300313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
Autoantibodies to chromatin and dsDNA are a hallmark of systemic lupus erythematosus (SLE). In a mouse model of monogenic human SLE caused by DNASE1L3 deficiency, the anti-DNA response is dependent on endosomal nucleic acid-sensing TLRs TLR7 and TLR9. In this study, we report that this response also required TLR2, a surface receptor for microbial products that is primarily expressed on myeloid cells. Cell transfers into lymphopenic DNASE1L3-deficient mice showed that TLR2 was required for anti-DNA Ab production by lymphocytes. TLR2 was detectably expressed on B cells and facilitated the production of IL-6 by B cells activated in the presence of microbial products. Accordingly, treatment with broad-spectrum antibiotics or Ab-mediated blockade of IL-6 delayed the anti-DNA response in DNASE1L3-deficient mice. These studies reveal an unexpected B cell-intrinsic role of TLR2 in systemic autoreactivity to DNA, and they suggest that microbial products may synergize with self-DNA in the activation of autoreactive B cells in SLE.
Collapse
Affiliation(s)
- Chetna Soni
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sohei Makita
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anna Eichinger
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU, Munich, Germany
| | - Lee Serpas
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Vanja Sisirak
- CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 33076 Bordeaux, France Dept. of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Boris Reizis
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
4
|
Carrillo-Rodríguez P, Robles-Guirado JÁ, Cruz-Palomares A, Palacios-Pedrero MÁ, González-Paredes E, Más-Ciurana A, Franco-Herrera C, Ruiz-de-Castroviejo-Teba PA, Lario A, Longobardo V, Montosa-Hidalgo L, Pérez-Sánchez-Cañete MM, Corzo-Corbera MM, Redondo-Sánchez S, Jodar AB, Blanco FJ, Zumaquero E, Merino R, Sancho J, Zubiaur M. Extracellular vesicles from pristane-treated CD38-deficient mice express an anti-inflammatory neutrophil protein signature, which reflects the mild lupus severity elicited in these mice. Front Immunol 2022; 13:1013236. [DOI: 10.3389/fimmu.2022.1013236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
In CD38-deficient (Cd38-/-) mice intraperitoneal injection of pristane induces a lupus-like disease, which is milder than that induced in WT mice, showing significant differences in the inflammatory and autoimmune processes triggered by pristane. Extracellular vesicles (EV) are present in all body fluids. Shed by cells, their molecular make-up reflects that of their cell of origin and/or tissue pathological situation. The aim of this study was to analyze the protein composition, protein abundance, and functional clustering of EV released by peritoneal exudate cells (PECs) in the pristane experimental lupus model, to identify predictive or diagnostic biomarkers that might discriminate the autoimmune process in lupus from inflammatory reactions and/or normal physiological processes. In this study, thanks to an extensive proteomic analysis and powerful bioinformatics software, distinct EV subtypes were identified in the peritoneal exudates of pristane-treated mice: 1) small EV enriched in the tetraspanin CD63 and CD9, which are likely of exosomal origin; 2) small EV enriched in CD47 and CD9, which are also enriched in plasma-membrane, membrane-associated proteins, with an ectosomal origin; 3) small EV enriched in keratins, ECM proteins, complement/coagulation proteins, fibrin clot formation proteins, and endopetidase inhibitor proteins. This enrichment may have an inflammation-mediated mesothelial-to-mesenchymal transition origin, representing a protein corona on the surface of peritoneal exudate EV; 4) HDL-enriched lipoprotein particles. Quantitative proteomic analysis allowed us to identify an anti-inflammatory, Annexin A1-enriched pro-resolving, neutrophil protein signature, which was more prominent in EV from pristane-treated Cd38-/- mice, and quantitative differences in the protein cargo of the ECM-enriched EV from Cd38-/- vs WT mice. These differences are likely to be related with the distinct inflammatory outcome shown by Cd38-/- vs WT mice in response to pristane treatment. Our results demonstrate the power of a hypothesis-free and data-driven approach to transform the heterogeneity of the peritoneal exudate EV from pristane-treated mice in valuable information about the relative proportion of different EV in a given sample and to identify potential protein markers specific for the different small EV subtypes, in particular those proteins defining EV involved in the resolution phase of chronic inflammation.
Collapse
|
5
|
Hsieh YT, Chou YC, Kuo PY, Tsai HW, Yen YT, Shiau AL, Wang CR. Down-regulated miR-146a expression with increased neutrophil extracellular traps and apoptosis formation in autoimmune-mediated diffuse alveolar hemorrhage. J Biomed Sci 2022; 29:62. [PMID: 36028828 PMCID: PMC9413930 DOI: 10.1186/s12929-022-00849-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing evidences have suggested an important role of microRNAs (miRNAs) in regulating cell death processes including NETosis and apoptosis. Dysregulated expression of miRNAs and increased formation of neutrophil extracellular traps (NETs) and apoptosis participate in autoimmune-mediated diffuse alveolar hemorrhage (DAH), mostly associated with pulmonary capillaritis in systemic lupus erythematosus (SLE) patients. In particular, besides the inhibition of apoptosis, miR-146a can control innate and acquired immune responses, and regulate the toll-like receptor pathway through targeting TRAF6 to reduce the expression of pro-inflammatory cytokines/chemokines like IL-8, a NETosis inducer. METHODS Expression of miR-146a, TRAF6 and NETs were examined in peripheral blood neutrophils (PBNs) and lung tissues from SLE-associated DAH patients, and in neutrophils and pristane-induced DAH lung tissues from C57BL/6 mice. To assess NETs formation, we examined NETosis-related DNAs morphology and crucial mediators including protein arginine deiminase 4 and citrullinated Histone 3. Expression of miR-146a and its endogenous RNA SNHG16 were studied in HL-60 promyelocytic cells and MLE-12 alveolar cells during NETosis and apoptosis processes, respectively. MiR-146a-overexpressed and CRISPR-Cas13d-mediated SNHG16-silenced HL-60 cells were investigated for NETosis. MiR-146a-overexpressed MLE-12 cells were analyzed for apoptosis. Pristane-injected mice received intra-pulmonary miR-146a delivery to evaluate therapeutic efficacy in DAH. RESULTS In DAH patients, there were down-regulated miR-146a levels with increased TRAF6 expression and PMA/LPS-induced NETosis in PBNs, and down-regulated miR-146a levels with increased TRAF6, high-mobility group box 1 (HMGB1), IL-8, NETs and apoptosis expression in lung tissues. HMGB1-stimulated mouse neutrophils had down-regulated miR-146a levels with increased TRAF6, IL-8 and NETs expression. PMA-stimulated HL-60 cells had down-regulated miR-146a levels with enhanced NETosis. MiR-146a-overexpressed or SNHG16-silenced HL-60 cells showed reduced NETosis. Apoptotic MLE-12 cells had down-regulated miR-146a expression and increased HMGB1 release, while miR-146a-overexpressed MLE-12 cells showed reduced apoptosis and HMGB1 production. There were down-regulated miR-146a levels with increased TRAF6, HMGB1, IL-8, NETs and apoptosis expression in mouse DAH lung tissues. Intra-pulmonary miR-146a delivery could suppress DAH by reducing TRAF6, IL-8, NETs and apoptosis expression. CONCLUSIONS Our results demonstrate firstly down-regulated pulmonary miR-146a levels with increased TRAF6 and IL-8 expression and NETs and apoptosis formation in autoimmune-mediated DAH, and implicate a therapeutic potential of intra-pulmonary miR-146a delivery.
Collapse
Affiliation(s)
- Yu-Tung Hsieh
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Pin-Yu Kuo
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Wen Tsai
- Departments of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Yen
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chrong-Reen Wang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Aschman T, Schaffer S, Biniaris Georgallis SI, Triantafyllopoulou A, Staeheli P, Voll RE. Interferon Lambda Regulates Cellular and Humoral Immunity in Pristane-Induced Lupus. Int J Mol Sci 2021; 22:ijms222111747. [PMID: 34769174 PMCID: PMC8584021 DOI: 10.3390/ijms222111747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
A pivotal role of type I interferons in systemic lupus erythematosus (SLE) is widely accepted. Type III interferons (IFN-λ) however, the most recently discovered cytokines grouped within the interferon family, have not been extensively studied in lupus disease models yet. Growing evidence suggests a role for IFN-λ in regulating both innate and adaptive immune responses, and increased serum concentrations have been described in multiple autoimmune diseases including SLE. Using the pristane-induced lupus model, we found that mice with defective IFN-λ receptors (Ifnlr1−/−) showed increased survival rates, decreased lipogranuloma formation and reduced anti-dsDNA autoantibody titers in the early phase of autoimmunity development compared to pristane-treated wild-type mice. Moreover, Ifnlr1−/− mice treated with pristane had reduced numbers of inflammatory mononuclear phagocytes and cNK cells in their kidneys, resembling untreated control mice. Systemically, circulating B cells and monocytes (CD115+Ly6C+) were reduced in pristane-treated Ifnlr1−/− mice. The present study supports a significant role for type III interferons in the pathogenesis of pristane-induced murine autoimmunity as well as in systemic and renal inflammation. Although the absence of type III interferon receptors does not completely prevent the development of autoantibodies, type III interferon signaling accelerates the development of autoimmunity and promotes a pro-inflammatory environment in autoimmune-prone hosts.
Collapse
Affiliation(s)
- Tom Aschman
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (S.S.); (S.I.B.G.); (A.T.)
- Department of Neuropathology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117 Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
- Correspondence: (T.A.); (R.E.V.)
| | - Sandra Schaffer
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (S.S.); (S.I.B.G.); (A.T.)
| | - Stylianos Iason Biniaris Georgallis
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (S.S.); (S.I.B.G.); (A.T.)
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (S.S.); (S.I.B.G.); (A.T.)
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg im Breisgau, Germany;
| | - Reinhard E. Voll
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; (S.S.); (S.I.B.G.); (A.T.)
- Center for Chronic Immunodeficiency (CCI), Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Correspondence: (T.A.); (R.E.V.)
| |
Collapse
|
7
|
Kiripolsky J, Kasperek EM, Zhu C, Li QZ, Wang J, Yu G, Kramer JM. Immune-Intrinsic Myd88 Directs the Production of Antibodies With Specificity for Extracellular Matrix Components in Primary Sjögren's Syndrome. Front Immunol 2021; 12:692216. [PMID: 34381449 PMCID: PMC8350326 DOI: 10.3389/fimmu.2021.692216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
Primary Sjögren's syndrome is an autoimmune disease that is predominantly seen in women. The disease is characterized by exocrine gland dysfunction in combination with serious systemic manifestations. At present, the causes of pSS are poorly understood. Pulmonary and renal inflammation are observed in pSS mice, reminiscent of a subset of pSS patients. A growing body of evidence indicates that inflammation mediated by Damage-Associated Molecular Patterns (DAMPs) contributes to autoimmunity, although this is not well-studied in pSS. Degraded extracellular matrix (ECM) constituents can serve as DAMPs by binding pattern-recognition receptors and activating Myd88-dependent signaling cascades, thereby exacerbating and perpetuating inflammatory cascades. The ECM components biglycan (Bgn) and decorin (Dcn) mediate sterile inflammation and both are implicated in autoimmunity. The objective of this study was to determine whether these ECM components and anti-ECM antibodies are altered in a pSS mouse model, and whether this is dependent on Myd88 activation in immune cells. Circulating levels of Bgn and Dcn were similar among pSS mice and controls and tissue expression studies revealed pSS mice had robust expression of both Bgn and Dcn in the salivary tissue, saliva, lung and kidney. Sera from pSS mice displayed increased levels of autoantibodies directed against ECM components when compared to healthy controls. Further studies using sera derived from conditional knockout pSS mice demonstrated that generation of these autoantibodies relies, at least in part, on Myd88 expression in the hematopoietic compartment. Thus, this study demonstrates that ECM degradation may represent a novel source of chronic B cell activation in the context of pSS.
Collapse
Affiliation(s)
- Jeremy Kiripolsky
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Eileen M. Kasperek
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Chengsong Zhu
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Quan-Zhen Li
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jia Wang
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Guan Yu
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jill M. Kramer
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
- Department of Oral Diagnostics Sciences, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| |
Collapse
|
8
|
Marks KE, Cho K, Stickling C, Reynolds JM. Toll-like Receptor 2 in Autoimmune Inflammation. Immune Netw 2021; 21:e18. [PMID: 34277108 PMCID: PMC8263214 DOI: 10.4110/in.2021.21.e18] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
TLR signaling is critical for broad scale immune recognition of pathogens and/or danger molecules. TLRs are particularly important for the activation and the maturation of cells comprising the innate immune response. In recent years it has become apparent that several different TLRs regulate the function of lymphocytes as well, albeit to a lesser degree compared to innate immunity. TLR2 heterodimerizes with either TLR1 or TLR6 to broadly recognize bacterial lipopeptides as well as several danger-associated molecular patterns. In general, TLR2 signaling promotes immune cell activation leading to tissue inflammation, which is advantageous for combating an infection. Conversely, inappropriate or dysfunctional TLR2 signaling leading to an overactive inflammatory response could be detrimental during sterile inflammation and autoimmune disease. This review will highlight and discuss recent research advances linking TLR2 engagement to autoimmune inflammation.
Collapse
Affiliation(s)
- Kathryne E Marks
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kaylin Cho
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Courtney Stickling
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joseph M Reynolds
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
9
|
Marques CPC, Rodrigues VP, de Carvalho LC, Nichilatti LP, Franco MM, Patrício FJB, Magalhães M, de Andrade MS, Benatti BB. Expression of Toll-like receptors 2 and 4 in the saliva of patients with systemic lupus erythematosus and chronic periodontitis. Clin Rheumatol 2021; 40:2727-2734. [PMID: 33570702 DOI: 10.1007/s10067-020-05560-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/08/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the expression of salivary Toll-like receptors (TRL) 2 and 4 in patients with systemic lupus erythematosus (SLE) and chronic periodontitis (CP). METHODS A case-control study was conducted with 77 participants (42 SLE and 35 non-SLE) stratified according to CP diagnosis criteria. Periodontal parameters consisted of clinical attachment level (CAL), probing depth (PD), the visible plaque index (VPI), and the gingival bleeding index (GBI). Salivary TRL 2 and 4 expressions were determined by quantitative real-time polymerase chain reaction (RT-PCR). Statistical analysis included Mann-Whitney U test, Kruskal-Wallis test, Spearman's correlation rank, and multiple linear regression. RESULTS Patients with isolated SLE or CP had higher TLR 2 and TLR 4 expression in their saliva samples (P < 0.05). The group with both SLE and CP had lower TLR 2 and 4 expressions (P < 0.05). TLR 2 and TLR 4 showed significant negative correlations with PD, CAL, and GBI in SLE patients, and a significant positive correlation with periodontal parameters in non-SLE patients. CP was independently associated with reduction of TLR2 and TLR4 expression, even after adjusting for clinical data and current drug use. CONCLUSION Reduced TRL 2 and 4 expression in saliva was associated with the presence of CP in SLE patients. Key Points • Patients affected by isolated CP or SLE had higher TLR2 and TLR4 expression. • TLR under-expression may be associated with a worse periodontal status in SLE. • Abnormalities in TLRs expression may increase the susceptibility to periodontitis.
Collapse
Affiliation(s)
- Consuelo P C Marques
- Dentistry Graduate Program, Federal University of Maranhão, São Luís, Maranhão, Brazil.,School of Medicine, Federal University of Maranhão, Pinheiro, Maranhão, Brazil
| | - Vandilson P Rodrigues
- Dentistry Graduate Program, Federal University of Maranhão, São Luís, Maranhão, Brazil. .,Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), President Dutra Hospital of the Federal University of Maranhão, São Luís, Maranhão, Brazil.
| | | | - Louise P Nichilatti
- School of Medicine, Federal University of Maranhão, Pinheiro, Maranhão, Brazil
| | - Mayra M Franco
- Dentistry Graduate Program, Federal University of Maranhão, São Luís, Maranhão, Brazil
| | - Fernando José B Patrício
- Laboratory of Genomic Studies and Histocompatibility, President Dutra Hospital of the Federal University of Maranhão, São Luís, Maranhão, Brazil
| | - Marcelo Magalhães
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), President Dutra Hospital of the Federal University of Maranhão, São Luís, Maranhão, Brazil
| | - Marcelo S de Andrade
- Laboratory of Genomic Studies and Histocompatibility, President Dutra Hospital of the Federal University of Maranhão, São Luís, Maranhão, Brazil
| | - Bruno B Benatti
- Dentistry Graduate Program, Federal University of Maranhão, São Luís, Maranhão, Brazil
| |
Collapse
|
10
|
McClung DM, Kalusche WJ, Jones KE, Ryan MJ, Taylor EB. Hypertension and endothelial dysfunction in the pristane model of systemic lupus erythematosus. Physiol Rep 2021; 9:e14734. [PMID: 33527772 PMCID: PMC7851437 DOI: 10.14814/phy2.14734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/19/2020] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases such as psoriasis, rheumatoid arthritis, and systemic lupus erythematosus (SLE) have high rates of hypertension and cardiovascular disease. Systemic lupus erythematosus is a prototypic autoimmune disorder that primarily affects women of childbearing age and is associated with a loss of self-tolerance, autoreactive B and T lymphocytes, and the production of autoantibodies, especially to nuclear components. In this study, we hypothesized that the pristane-inducible model of SLE would develop hypertension and vascular dysfunction as the disease progressed. To test this hypothesis, female C57BL/6 mice were administered PBS or pristane. Seven months after pristane administration, mice developed various autoantibodies, including anti-dsDNA IgG, anti-ssDNA IgG, and anti-nRNP IgG, as well as hypergammaglobulinemia. Several other immunological changes, including increased circulating neutrophils and increased CD4- CD8- (double negative) thymocytes were also detected. Mean arterial pressure (MAP) was elevated in pristane-treated mice when compared to PBS-treated mice. In addition, second-order mesenteric arteries from pristine-treated mice had impaired relaxation to the endothelium-dependent vasodilator acetylcholine compared to PBS-treated mice. These data suggest that the immune system dysfunction present in the pristane model of lupus contributes to the development of hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Daniel M. McClung
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - William J. Kalusche
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Katie E. Jones
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Michael J. Ryan
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
- G.V. (Sonny) Montgomery Veterans Affairs Medical CenterJacksonMSUSA
| | - Erin B. Taylor
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMSUSA
| |
Collapse
|
11
|
Mertowski S, Lipa P, Morawska I, Niedźwiedzka-Rystwej P, Bębnowska D, Hrynkiewicz R, Grywalska E, Roliński J, Załuska W. Toll-Like Receptor as a Potential Biomarker in Renal Diseases. Int J Mol Sci 2020; 21:ijms21186712. [PMID: 32933213 PMCID: PMC7554805 DOI: 10.3390/ijms21186712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 02/08/2023] Open
Abstract
One of the major challenges faced by modern nephrology is the identification of biomarkers associated with histopathological patterns or defined pathogenic mechanisms that may assist in the non-invasive diagnosis of kidney disease, particularly glomerulopathy. The identification of such molecules may allow prognostic subgroups to be established based on the type of disease, thereby predicting response to treatment or disease relapse. Advances in understanding the pathogenesis of diseases, such as membranous nephropathy, minimal change disease, focal segmental glomerulosclerosis, IgA (immunoglobulin A) nephropathy, and diabetic nephropathy, along with the progressive development and standardization of plasma and urine proteomics techniques, have facilitated the identification of an increasing number of molecules that may be useful for these purposes. The growing number of studies on the role of TLR (toll-like receptor) receptors in the pathogenesis of kidney disease forces contemporary researchers to reflect on these molecules, which may soon join the group of renal biomarkers and become a helpful tool in the diagnosis of glomerulopathy. In this article, we conducted a thorough review of the literature on the role of TLRs in the pathogenesis of glomerulopathy. The role of TLR receptors as potential marker molecules for the development of neoplastic diseases is emphasized more and more often, as prognostic factors in diseases on several epidemiological backgrounds.
Collapse
Affiliation(s)
- Sebastian Mertowski
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (J.R.)
- Correspondence: (S.M.); (P.N.-R.); (E.G.)
| | - Paulina Lipa
- Department of Genetics and Microbiology, Institute of Microbiology and Biotechnology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 St., 20-033 Lublin, Poland;
| | - Izabela Morawska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (J.R.)
| | - Paulina Niedźwiedzka-Rystwej
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (D.B.); (R.H.)
- Correspondence: (S.M.); (P.N.-R.); (E.G.)
| | - Dominika Bębnowska
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (D.B.); (R.H.)
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (D.B.); (R.H.)
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (J.R.)
- Correspondence: (S.M.); (P.N.-R.); (E.G.)
| | - Jacek Roliński
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (J.R.)
| | - Wojciech Załuska
- Department of Nephrology, Medical University of Lublin, 20-954 Lublin, Poland;
| |
Collapse
|
12
|
Agliano F, Karlinsey KS, Ragazzi M, Ménoret A, Vella AT. A benzimidazole inhibitor attenuates sterile inflammation induced in a model of systemic autoinflammation in female mice. Sci Rep 2020; 10:12100. [PMID: 32694575 PMCID: PMC7374700 DOI: 10.1038/s41598-020-68985-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Sterile stimuli can trigger inflammatory responses, and in some cases can lead to a variety of acute or chronic diseases. In this study, we hypothesize that a benzimidazole inhibitor may be used as a therapeutic in the treatment of sterile inflammation. In vitro, this inhibitor blocks TLR signalling and inflammatory responses. The benzimidazole inhibitor does not prevent mouse macrophage activation after stimulation with 2,6,10,14-tetramethylpentadecane (TMPD, also known as pristane), a hydrocarbon oil that mimics features of sterile inflammation when injected in vivo. However, C57BL/6J female mice treated with the benzimidazole inhibitor exhibited a significant reduction of pristane-dependent induction of splenocyte number and weight. Conversely, no significant difference was observed in males. Using mass spectrometry, we found that the urine of pristane-injected mice contained increased levels of putative markers for several inflammatory diseases, which were reduced by the benzimidazole inhibitor. To study the mechanism, we showed that pristane-injected mice had increased cell free DNA in serum, which was not impacted by inhibitor treatment. However, chemokine release (e.g. MCP-1, RANTES and TARC) was significantly reduced in inhibitor-treated mice. Thus, the benzimidazole inhibitor might be used as a new drug to block the recruitment of immune cells during sterile inflammatory diseases in humans.
Collapse
Affiliation(s)
- Federica Agliano
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Keaton S Karlinsey
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Michael Ragazzi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Antoine Ménoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
13
|
Peixoto TV, Carrasco S, Botte DAC, Catanozi S, Parra ER, Lima TM, Ugriumov N, Soriano FG, de Mello SBV, Rodrigues CM, Goldenstein-Schainberg C. CD4+CD69+ T cells and CD4+CD25+FoxP3+ Treg cells imbalance in peripheral blood, spleen and peritoneal lavage from pristane-induced systemic lupus erythematosus (SLE) mice. Adv Rheumatol 2019; 59:30. [DOI: 10.1186/s42358-019-0072-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/02/2019] [Indexed: 12/18/2022] Open
|
14
|
Soni C, Reizis B. Self-DNA at the Epicenter of SLE: Immunogenic Forms, Regulation, and Effects. Front Immunol 2019; 10:1601. [PMID: 31354738 PMCID: PMC6637313 DOI: 10.3389/fimmu.2019.01601] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Self-reactive B cells generated through V(D)J recombination in the bone marrow or through accrual of random mutations in secondary lymphoid tissues are mostly purged or edited to prevent autoimmunity. Yet, 10–20% of all mature naïve B cells in healthy individuals have self-reactive B cell receptors (BCRs). In patients with serologically active systemic lupus erythematosus (SLE) the percentage increases up to 50%, with significant self-DNA reactivity that correlates with disease severity. Endogenous or self-DNA has emerged as a potent antigen in several autoimmune disorders, particularly in SLE. However, the mechanism(s) regulating or preventing anti-DNA antibody production remain elusive. It is likely that in healthy subjects, DNA-reactive B cells avoid activation due to the unavailability of endogenous DNA, which is efficiently degraded through efferocytosis and various DNA-processing proteins. Genetic defects, physiological, and/or pathological conditions can override these protective checkpoints, leading to autoimmunity. Plausibly, increased availability of immunogenic self-DNA may be the key initiating event in the loss of tolerance of otherwise quiescent DNA-reactive B cells. Indeed, mutations impairing apoptotic cell clearance pathways and nucleic acid metabolism-associated genes like DNases, RNases, and their sensors are known to cause autoimmune disorders including SLE. Here we review the literature supporting the idea that increased availability of DNA as an immunogen or adjuvant, or both, may cause the production of pathogenic anti-DNA antibodies and subsequent manifestations of clinical disease such as SLE. We discuss the main cellular players involved in anti-DNA responses; the physical forms and sources of immunogenic DNA in autoimmunity; the DNA-protein complexes that render DNA immunogenic; the regulation of DNA availability by intracellular and extracellular DNases and the autoimmune pathologies associated with their dysfunction; the cytosolic and endosomal sensors of immunogenic DNA; and the cytokines such as interferons that drive auto-inflammatory and autoimmune pathways leading to clinical disease. We propose that prevention of DNA availability by aiding extracellular DNase activity could be a viable therapeutic modality in controlling SLE.
Collapse
Affiliation(s)
- Chetna Soni
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Boris Reizis
- Department of Pathology, New York University School of Medicine, New York, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Eichhorst A, Daniel C, Rzepka R, Sehnert B, Nimmerjahn F, Voll RE, Chevalier N. Relevance of Receptor for Advanced Glycation end Products (RAGE) in Murine Antibody-Mediated Autoimmune Diseases. Int J Mol Sci 2019; 20:ijms20133234. [PMID: 31266174 PMCID: PMC6651235 DOI: 10.3390/ijms20133234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 12/24/2022] Open
Abstract
It is incompletely understood how self-antigens become targets of humoral immunity in antibody-mediated autoimmune diseases. In this context, alarmins are discussed as an important level of regulation. Alarmins are recognized by various receptors, such as receptor for advanced glycation end products (RAGE). As RAGE is upregulated under inflammatory conditions, strongly binds nucleic acids and mediates pro-inflammatory responses upon alarmin recognition, our aim was to examine its contribution to immune complex-mediated autoimmune diseases. This question was addressed employing RAGE−/− animals in murine models of pristane-induced lupus, collagen-induced, and serum-transfer arthritis. Autoantibodies were assessed by enzyme-linked immunosorbent assay, renal disease by quantification of proteinuria and histology, arthritis by scoring joint inflammation. The associated immune status was determined by flow cytometry. In both disease entities, we detected tendentiously decreased autoantibody levels in RAGE−/− mice, however no differences in clinical outcome. In accordance with autoantibody levels, a subgroup of the RAGE−/− animals showed a decrease in plasma cells, and germinal center B cells and an increase in follicular B cells. Based on our results, we suggest that RAGE deficiency alone does not significantly affect antibody-mediated autoimmunity. RAGE may rather exert its effects along with other receptors linking environmental factors to auto-reactive immune responses.
Collapse
Affiliation(s)
- Alexandra Eichhorst
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, 91054 Erlangen, Bavaria, Germany
| | - Rita Rzepka
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Falk Nimmerjahn
- Department of Biology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, 91058 Erlangen, Bavaria, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany
| | - Nina Chevalier
- Department of Rheumatology and Clinical Immunology, Medical Centre-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Baden-Wuerttemberg, Germany.
| |
Collapse
|
16
|
Ning F, Li X, Yu L, Zhang B, Zhao Y, Liu Y, Zhao B, Shang Y, Hu X. Hes1 attenuates type I IFN responses via VEGF-C and WDFY1. J Exp Med 2019; 216:1396-1410. [PMID: 31015298 PMCID: PMC6547865 DOI: 10.1084/jem.20180861] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/22/2018] [Accepted: 04/04/2019] [Indexed: 12/26/2022] Open
Abstract
Transcription factor Hes1 acts as a homeostatic negative regulator of type I interferon production to restrain interferon-mediated immune responses, including antiviral immunity and autoimmune conditions. Mechanistically, Hes1 suppresses interferon expression by targeting a regulatory circuit composed of WDFY1 and VEGF-C. Induction of type I interferons (IFNs) is critical for eliciting competent immune responses, especially antiviral immunity. However, uncontrolled IFN production contributes to pathogenesis of autoimmune and inflammatory diseases. We found that transcription factor Hes1 suppressed production of type I IFNs and expression of IFN-stimulated genes. Functionally, Hes1-deficient mice displayed a heightened IFN signature in vivo, mounted enhanced resistance against encephalomyocarditis virus infection, and showed signs of exacerbated experimental lupus nephritis. Mechanistically, Hes1 did not suppress IFNs via direct transcriptional repression of IFN-encoding genes. Instead, Hes1 attenuated activation of TLR upstream signaling by inhibition of an adaptor molecule, WDFY1. Genome-wide assessment of Hes1 occupancy revealed that suppression of WDFY1 was secondary to direct binding and thus enhancement of expression of VEGF-C by Hes1, making Vegfc a rare example of an Hes1 positively regulated gene. In summary, these results identified Hes1 as a homeostatic negative regulator of type I IFNs for the maintenance of immune balance in the context of antiviral immunity and autoimmune diseases.
Collapse
Affiliation(s)
- Fei Ning
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Xiaoyu Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Li Yu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Bin Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Yuna Zhao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control & Prevention, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Yu Liu
- State Key Laboratory of Virology, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, China
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Yingli Shang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control & Prevention, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China .,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
17
|
van Dam LS, Rabelink TJ, van Kooten C, Teng YKO. Clinical Implications of Excessive Neutrophil Extracellular Trap Formation in Renal Autoimmune Diseases. Kidney Int Rep 2018; 4:196-211. [PMID: 30775617 PMCID: PMC6365354 DOI: 10.1016/j.ekir.2018.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/02/2018] [Accepted: 11/12/2018] [Indexed: 12/20/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular DNA structures covered with antimicrobial peptides, danger molecules, and autoantigens that can be released by neutrophils. NETs are an important first-line defense mechanism against bacterial, viral, fungal, and parasitic infections, but they can also play a role in autoimmune diseases. NETs are immunogenic and toxic structures that are recognized by the autoantibodies of patients with antineutrophil cytoplasmic antibodies−associated vasculitis (AAV) (i.e., against myeloperoxidase or proteinase-3) and systemic lupus erythematosus (SLE) (i.e., against double-stranded DNA, histones, or nucleosomes). There is cumulating preclinical and clinical evidence that both excessive formation and impaired degradation of NETs are involved in the pathophysiology of AAV and SLE. These autoimmune diseases give rise to 2 clinically and pathologically distinct forms of glomerulonephritis (GN), respectively, crescentic pauci-immune GN and immune complex−mediated GN. Therefore, it is relevant to understand the different roles NET formation can play in the pathophysiology of these most prevalent renal autoimmune diseases. This review summarizes the current concepts on the role of NET formation in the pathophysiology of AAV and SLE, and provides a translational perspective on the clinical implications of NETs, such as potential therapeutic approaches that target NET formation in these renal autoimmune diseases.
Collapse
Affiliation(s)
- Laura S van Dam
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Cees van Kooten
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Y K Onno Teng
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
18
|
Rizzetto L, Fava F, Tuohy KM, Selmi C. Connecting the immune system, systemic chronic inflammation and the gut microbiome: The role of sex. J Autoimmun 2018; 92:12-34. [PMID: 29861127 DOI: 10.1016/j.jaut.2018.05.008] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Unresolved low grade systemic inflammation represents the underlying pathological mechanism driving immune and metabolic pathways involved in autoimmune diseases (AID). Mechanistic studies in animal models of AID and observational studies in patients have found alterations in gut microbiota communities and their metabolites, suggesting a microbial contribution to the onset or progression of AID. The gut microbiota and its metabolites have been shown to influence immune functions and immune homeostasis both within the gut and systematically. Microbial derived-short chain fatty acid (SCFA) and bio-transformed bile acid (BA) have been shown to influence the immune system acting as ligands specific cell signaling receptors like GPRCs, TGR5 and FXR, or via epigenetic processes. Similarly, intestinal permeability (leaky gut) and bacterial translocation are important contributors to chronic systemic inflammation and, without repair of the intestinal barrier, might represent a continuous inflammatory stimulus capable of triggering autoimmune processes. Recent studies indicate gender-specific differences in immunity, with the gut microbiota shaping and being concomitantly shaped by the hormonal milieu governing differences between the sexes. A bi-directional cross-talk between microbiota and the endocrine system is emerging with bacteria being able to produce hormones (e.g. serotonin, dopamine and somatostatine), respond to host hormones (e.g. estrogens) and regulate host hormones' homeostasis (e.g by inhibiting gene prolactin transcription or converting glucocorticoids to androgens). We review herein how gut microbiota and its metabolites regulate immune function, intestinal permeability and possibly AID pathological processes. Further, we describe the dysbiosis within the gut microbiota observed in different AID and speculate how restoring gut microbiota composition and its regulatory metabolites by dietary intervention including prebiotics and probiotics could help in preventing or ameliorating AID. Finally, we suggest that, given consistent observations of microbiota dysbiosis associated with AID and the ability of SCFA and BA to regulate intestinal permeability and inflammation, further mechanistic studies, examining how dietary microbiota modulation can protect against AID, hold considerable potential to tackle increased incidence of AID at the population level.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy.
| | - Francesca Fava
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Kieran M Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Italy; BIOMETRA Department, University of Milan, Italy
| |
Collapse
|
19
|
Biermann MHC, Boeltz S, Pieterse E, Knopf J, Rech J, Bilyy R, van der Vlag J, Tincani A, Distler JHW, Krönke G, Schett GA, Herrmann M, Muñoz LE. Autoantibodies Recognizing Secondary NEcrotic Cells Promote Neutrophilic Phagocytosis and Identify Patients With Systemic Lupus Erythematosus. Front Immunol 2018; 9:989. [PMID: 29867966 PMCID: PMC5949357 DOI: 10.3389/fimmu.2018.00989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/20/2018] [Indexed: 12/15/2022] Open
Abstract
Deficient clearance of apoptotic cells reportedly contributes to the etiopathogenesis of the autoimmune disease systemic lupus erythematosus (SLE). Based on this knowledge, we developed a highly specific and sensitive test for the detection of SLE autoantibodies (AAb) utilizing secondary NEcrotic cell (SNEC)-derived material as a substrate. The goal of the present study was to validate the use of SNEC as an appropriate antigen for the diagnosis of SLE in large cohort of patients. We confirmed the presence of apoptotically modified autoantigens on SNEC (dsDNA, high mobility group box 1 protein, apoptosis-associated chromatin modifications, e.g., histones H3-K27-me3; H2A/H4 AcK8,12,16; and H2B-AcK12). Anti-SNEC AAb were measured in the serum of 155 patients with SLE, 89 normal healthy donors (NHD), and 169 patients with other autoimmune connective tissue diseases employing SNEC-based indirect enzyme-linked immunosorbent assay (SNEC ELISA). We compared the test performance of SNEC ELISA with the routine diagnostic tests dsDNA Farr radioimmunoassay (RIA) and nucleosome-based ELISA (anti-dsDNA-NcX-ELISA). SNEC ELISA distinguished patients with SLE with a specificity of 98.9% and a sensitivity of 70.6% from NHD clearly surpassing RIA and anti-dsDNA-NcX-ELISA. In contrast to the other tests, SNEC ELISA significantly discriminated patients with SLE from patients with rheumatoid arthritis, primary anti-phospholipid syndrome, spondyloarthropathy, psoriatic arthritis, and systemic sclerosis. A positive test result in SNEC ELISA significantly correlated with serological variables and reflected the uptake of opsonized SNEC by neutrophils. This stresses the relevance of SNECs in the pathogenesis of SLE. We conclude that SNEC ELISA allows for the sensitive detection of pathologically relevant AAb, enabling its diagnostic usage. A positive SNEC test reflects the opsonization of cell remnants by AAb, the neutrophil recruitment to tissues, and the enhancement of local and systemic inflammatory responses.
Collapse
Affiliation(s)
- Mona H C Biermann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sebastian Boeltz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Elmar Pieterse
- Department of Nephrology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jasmin Knopf
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rostyslav Bilyy
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Angela Tincani
- Division of Rheumatology and Clinical Immunology, Department of Clinical and Experimental Sciences, Spedali Civili and University of Brescia, Brescia, Italy
| | - Jörg H W Distler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Andreas Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
20
|
Schell SL, Soni C, Fasnacht MJ, Domeier PP, Cooper TK, Rahman ZSM. Mer Receptor Tyrosine Kinase Signaling Prevents Self-Ligand Sensing and Aberrant Selection in Germinal Centers. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:4001-4015. [PMID: 29118245 DOI: 10.4049/jimmunol.1700611] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 10/11/2017] [Indexed: 04/04/2025]
Abstract
Mer tyrosine kinase (Mer) signaling maintains immune tolerance by clearing apoptotic cells (ACs) and inducing immunoregulatory signals. We previously showed that Mer-deficient mice (Mer-/-) have increased germinal center (GC) responses, T cell activation, and AC accumulation within GCs. Accumulated ACs in GCs can undergo necrosis and release self-ligands, which may influence the outcome of a GC response and selection. In this study, we generated Mer-/- mice with a global MyD88, TLR7, or TLR9 deficiency and cell type-specific MyD88 deficiency to study the functional correlation between Mer and TLRs in the development of GC responses and autoimmunity. We found that GC B cell-intrinsic sensing of self-RNA, but not self-DNA, released from dead cells accumulated in GCs drives enhanced GC responses in Mer-/- mice. Although self-ligands directly affect GC B cell responses, the loss of Mer in dendritic cells promotes enhanced T cell activation and proinflammatory cytokine production. To study the impact of Mer deficiency on the development of autoimmunity, we generated autoimmune-prone B6.Sle1b mice deficient in Mer (Sle1bMer-/-). We observed accelerated autoimmunity development even under conditions where Sle1bMer-/- mice did not exhibit increased AC accumulation in GCs compared with B6.Sle1b mice, indicating that Mer immunoregulatory signaling in APCs regulates B cell selection and autoimmunity. We further found significant expansion, retention, and class-switching of autoreactive B cells in GCs under conditions where ACs accumulated in GCs of Sle1bMer-/- mice. Altogether, both the phagocytic and immunomodulatory functions of Mer regulate GC responses to prevent the development of autoimmunity.
Collapse
Affiliation(s)
- Stephanie L Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Chetna Soni
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Melinda J Fasnacht
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Phillip P Domeier
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Timothy K Cooper
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| |
Collapse
|
21
|
Mu Q, Kirby J, Reilly CM, Luo XM. Leaky Gut As a Danger Signal for Autoimmune Diseases. Front Immunol 2017; 8:598. [PMID: 28588585 PMCID: PMC5440529 DOI: 10.3389/fimmu.2017.00598] [Citation(s) in RCA: 372] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/05/2017] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelial lining, together with factors secreted from it, forms a barrier that separates the host from the environment. In pathologic conditions, the permeability of the epithelial lining may be compromised allowing the passage of toxins, antigens, and bacteria in the lumen to enter the blood stream creating a “leaky gut.” In individuals with a genetic predisposition, a leaky gut may allow environmental factors to enter the body and trigger the initiation and development of autoimmune disease. Growing evidence shows that the gut microbiota is important in supporting the epithelial barrier and therefore plays a key role in the regulation of environmental factors that enter the body. Several recent reports have shown that probiotics can reverse the leaky gut by enhancing the production of tight junction proteins; however, additional and longer term studies are still required. Conversely, pathogenic bacteria that can facilitate a leaky gut and induce autoimmune symptoms can be ameliorated with the use of antibiotic treatment. Therefore, it is hypothesized that modulating the gut microbiota can serve as a potential method for regulating intestinal permeability and may help to alter the course of autoimmune diseases in susceptible individuals.
Collapse
Affiliation(s)
- Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Jay Kirby
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | | | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
22
|
Kienhöfer D, Hahn J, Stoof J, Csepregi JZ, Reinwald C, Urbonaviciute V, Johnsson C, Maueröder C, Podolska MJ, Biermann MH, Leppkes M, Harrer T, Hultqvist M, Olofsson P, Munoz LE, Mocsai A, Herrmann M, Schett G, Holmdahl R, Hoffmann MH. Experimental lupus is aggravated in mouse strains with impaired induction of neutrophil extracellular traps. JCI Insight 2017; 2:92920. [PMID: 28515366 DOI: 10.1172/jci.insight.92920] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 11/17/2022] Open
Abstract
Many effector mechanisms of neutrophils have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). Neutrophil extracellular traps (NETs) have been assigned a particularly detrimental role. Here we investigated the functional impact of neutrophils and NETs on a mouse model of lupus triggered by intraperitoneal injection of the cell death-inducing alkane pristane. Pristane-induced lupus (PIL) was aggravated in 2 mouse strains with impaired induction of NET formation, i.e., NOX2-deficient (Ncf1-mutated) and peptidyl arginine deiminase 4-deficient (PAD4-deficient) mice, as seen from elevated levels of antinuclear autoantibodies (ANAs) and exacerbated glomerulonephritis. We observed a dramatically reduced ability to form pristane-induced NETs in vivo in both Ncf1-mutated and PAD4-deficient mice, accompanied by higher levels of inflammatory mediators in the peritoneum. Similarly, neutropenic Mcl-1ΔMyelo mice exhibited higher levels of ANAs, which indicates a regulatory function in lupus of NETs and neutrophils. Blood neutrophils from Ncf1-mutated and human individuals with SLE exhibited exuberant spontaneous NET formation. Treatment with specific chemical NOX2 activators induced NET formation and ameliorated PIL. Our findings suggest that aberrant NET is one of the factors promoting experimental lupus-like autoimmunity by uncontrolled release of inflammatory mediators.
Collapse
Affiliation(s)
- Deborah Kienhöfer
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jonas Hahn
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Stoof
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janka Zsófia Csepregi
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christiane Reinwald
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Vilma Urbonaviciute
- Section of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Christian Maueröder
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Malgorzata J Podolska
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mona H Biermann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Moritz Leppkes
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Harrer
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | - Luis E Munoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Attila Mocsai
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rikard Holmdahl
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus H Hoffmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
23
|
Zhou S, Wang Y, Meng Y, Xiao C, Liu Z, Brohawn P, Higgs BW, Jallal B, Jia Q, Qu B, Huang X, Tang Y, Yao Y, Harley JB, Shen N. In Vivo Therapeutic Success of MicroRNA-155 Antagomir in a Mouse Model of Lupus Alveolar Hemorrhage. Arthritis Rheumatol 2016; 68:953-64. [PMID: 26556607 DOI: 10.1002/art.39485] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/22/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Diffuse alveolar hemorrhage (DAH) is a rare but life-threatening complication of systemic lupus erythematosus (SLE). Pristane-treated B6 mice develop severe DAH within 2 weeks of treatment. MicroRNA-155 (miR-155) is a pleiotropic microRNA that plays a crucial role in the regulation of immune responses. Recent studies have revealed a pathogenic role of miR-155 in various autoimmune disorders. The purpose of this study was to examine the role of miR-155 in the development of DAH in pristane-induced lupus using miR-155-knockout (miR-155(-/-)) mice and miR-155 antagomir to silence miR-155. METHODS DAH was induced by an intraperitoneal injection of 0.5 ml of pristane. MicroRNA-155 antagomir was administered intravenously to silence miR-155 expression. Lung tissues were collected for RNA extraction and were embedded in paraffin for sectioning. Gene expression profiling data were analyzed using Ingenuity Pathway Analysis. Real-time quantitative polymerase chain reaction analysis was used for single-gene validation. Luciferase reporter assay and argonaute 2 immunoprecipitation were performed for target validation. RESULTS MicroRNA-155 expression was significantly increased during the development of DAH. Disease progression was reduced in miR-155(-/-) mice as well as by in vivo silencing of miR-155 using a miR-155 antagomir. MicroRNA-155 silencing dampened pristane-induced ectopic activation of multiple inflammatory pathways and reduced the expression of proinflammatory cytokines. Several negative regulators of NF-κB signaling were inhibited by pristane and were reactivated in miR-155(-/-) mice. In particular, the antiinflammatory factor peroxisome proliferator-activated receptor α was identified as a direct target of miR-155. CONCLUSION MicroRNA-155 promotes pristane-induced lung inflammation. It contributes to ectopic activation of NF-κB signaling pathways by targeting multiple negative regulators. MicroRNA-155 antagomir may be a promising therapeutic strategy for treating acute lung inflammation in lupus.
Collapse
Affiliation(s)
- Shiyu Zhou
- Institute of Health Sciences of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Institute of Health Sciences of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Meng
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyuan Xiao
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | - Qian Jia
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Qu
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinfang Huang
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanjia Tang
- Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - John B Harley
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, and Cincinnati VA Medical Center, Cincinnati, Ohio
| | - Nan Shen
- Renji Hospital, Institute of Health Sciences of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
24
|
PÉREZ-FERRO MARÍA, SERRANO DEL CASTILLO CRISTINA, SÁNCHEZ-PERNAUTE OLGA. Cell Membrane-bound TLR2 and TLR4: Potential Predictors of Active Systemic Lupus Erythematosus and Lupus Nephritis. J Rheumatol 2016; 43:1444-5. [DOI: 10.3899/jrheum.151386] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Ebihara S, Tajima H, Ono M. Nuclear factor erythroid 2-related factor 2 is a critical target for the treatment of glucocorticoid-resistant lupus nephritis. Arthritis Res Ther 2016; 18:139. [PMID: 27301376 PMCID: PMC4908698 DOI: 10.1186/s13075-016-1039-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/31/2016] [Indexed: 12/25/2022] Open
Abstract
Background Dimethyl fumarate (DMF), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, has been proven effective for the systemic treatment of multiple sclerosis. The aim of this study is to evaluate the anti-inflammatory effects of Nrf2 activators on human renal mesangial cells (HRMCs) and the development of lupus nephritis (LN) in mice. Methods To assess Nrf2 activation in vitro, HRMCs were treated with safe doses of Nrf2 activators and prednisolone. The expression levels of Nrf2 and its target genes were measured using quantitative reverse transcription PCR and enzyme-linked immunosorbent assay. The anti-inflammatory effects of these compounds were assessed by measuring tumor necrosis factor alpha-induced cytokine secretion. Experimental LN was induced in female BALB/c mice by a single intraperitoneal injection of pristane. The urine albumin-to-creatinine ratio was measured at 20 weeks after injection. Pathological changes as well as protein and mRNA expression levels were assessed in the kidney obtained at the experimental end point. Oral administration of DMF or prednisolone to these mice was initiated after pristane injection. Results Nrf2 activators such as sulforaphane and DMF showed anti-inflammatory effects in HRMCs, whereas glucocorticoid (prednisolone) showed partial effects. Moreover, DMF ameliorated the development of kidney diseases in pristane-induced LN mice, whereas glucocorticoid had no effect. Conclusions Nrf2 activators showed stronger anti-inflammatory and organ-protective effects than glucocorticoid in the kidney. Thus, Nrf2 activators are potential therapeutic targets in glucocorticoid-resistant LN in humans.
Collapse
Affiliation(s)
- Shin Ebihara
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, Miyagi, 980-8575, Japan.
| | - Hideaki Tajima
- Department of Clinical laboratory, National Hospital Organization Mito Medical Center, 280 Sakuranosato, Ibaraki, higashi-ibaraki, Ibaraki, 311-3193, Japan
| | - Masao Ono
- Department of Clinical laboratory, National Hospital Organization Mito Medical Center, 280 Sakuranosato, Ibaraki, higashi-ibaraki, Ibaraki, 311-3193, Japan
| |
Collapse
|
26
|
Mahajan A, Herrmann M, Muñoz LE. Clearance Deficiency and Cell Death Pathways: A Model for the Pathogenesis of SLE. Front Immunol 2016; 7:35. [PMID: 26904025 PMCID: PMC4745266 DOI: 10.3389/fimmu.2016.00035] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/24/2016] [Indexed: 12/21/2022] Open
Abstract
Alterations of cell death pathways, including apoptosis and the neutrophil specific kind of death called NETosis, can represent a potential source of autoantigens. Defects in the clearance of apoptotic cells may be responsible for the initiation of systemic autoimmunity in several chronic inflammatory diseases, including systemic lupus erythematosus (SLE). Autoantigens are released mainly from secondary necrotic cells because of a defective clearance of apoptotic cells or an inefficient degradation of DNA-containing neutrophil extracellular traps (NETs). These modified autoantigens are presented by follicular dendritic cells to autoreactive B cells in germinal centers of secondary lymphoid organs. This results in the loss of self-tolerance and production of autoantibodies, a unifying feature of SLE. Immune complexes (IC) are formed from autoantibodies bound to uncleared cellular debris in blood or tissues. Clearance of IC by blood phagocytes, macrophages, and dendritic cells leads to proinflammatory cytokine secretion. In particular, plasmacytoid dendritic cells produce high amounts of interferon-α upon IC uptake, thereby contributing to the interferon signature of patients with SLE. The clearance of antinuclear IC via Fc-gamma receptors is considered a central event in amplifying inflammatory immune responses in SLE. Along with this, the accumulation of cell remnants represents an initiating event of the etiology, while the subsequent generation of autoantibodies against nuclear antigens (including NETs) results in the perpetuation of inflammation and tissue damage in patients with SLE. Here, we discuss the implications of defective clearance of apoptotic cells and NETs in the development of clinical manifestations in SLE.
Collapse
Affiliation(s)
- Aparna Mahajan
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| | - Luis E Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology , Erlangen , Germany
| |
Collapse
|
27
|
Dieker J, Tel J, Pieterse E, Thielen A, Rother N, Bakker M, Fransen J, Dijkman HBPM, Berden JH, de Vries JM, Hilbrands LB, van der Vlag J. Circulating Apoptotic Microparticles in Systemic Lupus Erythematosus Patients Drive the Activation of Dendritic Cell Subsets and Prime Neutrophils for NETosis. Arthritis Rheumatol 2016; 68:462-72. [PMID: 26360137 DOI: 10.1002/art.39417] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 08/27/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Circulating chromatin-containing apoptotic material and/or neutrophil extracellular traps (NETs) have been proposed to be an important driving force for the antichromatin autoimmune response in patients with systemic lupus erythematosus (SLE). The aim of this study was to determine the exact nature of microparticles in the circulation of SLE patients and to assess the effects of the microparticles on the immune system. METHODS We analyzed microparticles isolated from the plasma of patients with SLE, rheumatoid arthritis (RA), and systemic sclerosis (SSc), as well as from healthy subjects. The effects of the microparticles on blood-derived dendritic cells (DCs) and neutrophils were assessed by flow cytometry, enzyme-linked immunosorbent assay, and immunofluorescence microscopy. RESULTS In SLE patients, we identified microparticles that were highly positive for annexin V and apoptosis-modified chromatin that were not present in healthy subjects or in RA or SSc patients. These microparticles were mostly CD31+/CD45- (endothelial), partly CD45+/CD66b+ (granulocyte), and negative for B and T cell markers. Microparticles isolated from the plasma of SLE patients increased the expression of the costimulatory surface molecules CD40, CD80, CD83, and CD86 and the production of proinflammatory cytokines interleukin-6, tumor necrosis factor, and interferon-α by blood-derived plasmacytoid DCs (PDCs) and myeloid DCs (MDCs). SLE microparticles also primed blood-derived neutrophils for NETosis. Microparticles from healthy subjects and from RA or SSc patients exhibited no significant effects on MDCs, PDCs, and NETosis. CONCLUSION Circulating microparticles in SLE patients include a population of apoptotic cell-derived microparticles that has proinflammatory effects on PDCs and MDCs and enhances NETosis. These results underline the important role of apoptotic microparticles in driving the autoimmune response in SLE patients.
Collapse
Affiliation(s)
- Jürgen Dieker
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurjen Tel
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elmar Pieterse
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Astrid Thielen
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nils Rother
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marinka Bakker
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jaap Fransen
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Jo H Berden
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
28
|
Kimber I, Carrillo JC. Oral exposure to mineral oils: Is there an association with immune perturbation and autoimmunity? Toxicology 2016; 344-346:19-25. [DOI: 10.1016/j.tox.2016.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/19/2016] [Accepted: 01/22/2016] [Indexed: 12/18/2022]
|
29
|
Marsman G, Stephan F, de Leeuw K, Bulder I, Ruinard JT, de Jong J, Westra J, Bultink IE, Voskuyl AE, Aarden LA, Luken BM, Kallenberg CG, Zeerleder S. FSAP-mediated nucleosome release from late apoptotic cells is inhibited by autoantibodies present in SLE. Eur J Immunol 2015; 46:762-71. [DOI: 10.1002/eji.201546010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/19/2015] [Accepted: 11/26/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Gerben Marsman
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Femke Stephan
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Karina de Leeuw
- Department of Rheumatology and Clinical Immunology; University Medical Center; University of Groningen; Groningen The Netherlands
| | - Ingrid Bulder
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Jessica T. Ruinard
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Jan de Jong
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Johanna Westra
- Department of Rheumatology and Clinical Immunology; University Medical Center; University of Groningen; Groningen The Netherlands
| | - Irene E.M. Bultink
- Department of Rheumatology; VU University Medical Center; Amsterdam The Netherlands
| | - Alexandre E. Voskuyl
- Department of Rheumatology; VU University Medical Center; Amsterdam The Netherlands
| | - Lucien A. Aarden
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Brenda M. Luken
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Cees G.M. Kallenberg
- Department of Rheumatology and Clinical Immunology; University Medical Center; University of Groningen; Groningen The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology; Sanquin Research; Amsterdam; The Netherlands, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
- Department of Hematology; Academic Medical Centre; Amsterdam The Netherlands
| |
Collapse
|
30
|
Mu Q, Zhang H, Luo XM. SLE: Another Autoimmune Disorder Influenced by Microbes and Diet? Front Immunol 2015; 6:608. [PMID: 26648937 PMCID: PMC4663251 DOI: 10.3389/fimmu.2015.00608] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease. Despite years of study, the etiology of SLE is still unclear. Both genetic and environmental factors have been implicated in the disease mechanisms. In the past decade, a growing body of evidence has indicated an important role of gut microbes in the development of autoimmune diseases, including type 1 diabetes, rheumatoid arthritis, and multiple sclerosis. However, such knowledge on SLE is little, though we have already known that environmental factors can trigger the development of lupus. Several recent studies have suggested that alterations of the gut microbial composition may be correlated with SLE disease manifestations, while the exact roles of either symbiotic or pathogenic microbes in this disease remain to be explored. Elucidation of the roles of gut microbes - as well as the roles of diet that can modulate the composition of gut microbes - in SLE will shed light on how this autoimmune disorder develops, and provide opportunities for improved biomarkers of the disease and the potential to probe new therapies. In this review, we aim to compile the available evidence on the contributions of diet and gut microbes to SLE occurrence and pathogenesis.
Collapse
Affiliation(s)
- Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia Tech , Blacksburg, VA , USA
| | - Husen Zhang
- Department of Civil and Environmental Engineering, Virginia Tech , Blacksburg, VA , USA
| | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, Virginia Tech , Blacksburg, VA , USA
| |
Collapse
|
31
|
Roles of B Cell-Intrinsic TLR Signals in Systemic Lupus Erythematosus. Int J Mol Sci 2015; 16:13084-105. [PMID: 26068236 PMCID: PMC4490487 DOI: 10.3390/ijms160613084] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/31/2015] [Accepted: 06/03/2015] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a large family of pattern recognition receptors. TLR signals are involved in the pathogenesis of systemic lupus erythematosus. Mouse and human B cells constitutively express most TLRs. Many B cell subpopulations are highly responsive to certain TLR ligation, including B-1 B cells, transitional B cells, marginal zone B cells, germinal center B cell and memory B cells. The B cell-intrinsic TLR signals play critical roles during lupus process. In this review, roles of B cell-intrinsic TLR2, 4, 7, 8 and 9 signals are discussed during lupus pathogenesis in both mouse model and patients. Moreover, mechanisms underlying TLR ligation-triggered B cell activation and signaling pathways are highlighted.
Collapse
|
32
|
Wu Y, Zhang F, Ma J, Zhang X, Wu L, Qu B, Xia S, Chen S, Tang Y, Shen N. Association of large intergenic noncoding RNA expression with disease activity and organ damage in systemic lupus erythematosus. Arthritis Res Ther 2015; 17:131. [PMID: 25994030 PMCID: PMC4440330 DOI: 10.1186/s13075-015-0632-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 04/20/2015] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Despite growing evidence that large intergenic noncoding RNAs (lincRNAs) can regulate gene expression and widely take part in normal physiological and disease conditions, our knowledge of systemic lupus erythematosus (SLE)-related lincRNAs remains limited. The aim of this study was to detect the levels of four lincRNAs (ENST00000500949: linc0949, ENST00000500597: linc0597, ENST00000501992: linc1992, and ENST00000523995: linc3995) involved in innate immunity in the peripheral blood mononuclear cells (PBMCs) of patients with SLE and correlate these lincRNA levels with disease activity, organ damage, clinical features and medical therapies. METHODS PBMCs were obtained from 102 patients with SLE, 54 patients with rheumatoid arthritis (RA) and 76 healthy donors. lincRNA expression levels were measured by real-time quantitative polymerase chain reaction. Disease activity was assessed using the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) scores, and organ damage was evaluated with the Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index. RESULTS linc0949 and linc0597 were significantly decreased in patients with SLE compared with patients with RA and healthy control subjects. linc0949 was correlated with SLEDAI-2K score (r = -0.329, P = 0.0007), as well as with complement component C3 level (r = 0.348, P = 0.0003). The level of linc0949 was also reduced in patients with SLE who had the presence of cumulative organ damage. In addition, decreasing expression of linc0949 was associated with lupus nephritis. linc0949 expression significantly increased after treatment, whereas neither disease activity nor organ damage correlated with linc0597 expression. CONCLUSIONS Our results provide novel empirical evidence that linc0949 could be a potential biomarker for diagnosis, disease activity and therapeutic response in SLE.
Collapse
Affiliation(s)
- Yanfang Wu
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Feifei Zhang
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Yue Yang Road, Shanghai, 200031, People's Republic of China.
| | - Jianyang Ma
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Yue Yang Road, Shanghai, 200031, People's Republic of China.
| | - Xiaoyan Zhang
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Lingling Wu
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Bo Qu
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Shiwei Xia
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Shunle Chen
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
| | - Nan Shen
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shan Dong Middle Road, Shanghai, 200001, People's Republic of China.
- Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Yue Yang Road, Shanghai, 200031, People's Republic of China.
- Division of Rheumatology and the Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
33
|
Lupus nephritis: update on mechanisms of systemic autoimmunity and kidney immunopathology. Curr Opin Nephrol Hypertens 2014; 23:211-7. [PMID: 24662982 DOI: 10.1097/01.mnh.0000444816.57378.21] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Traditionally, lupus nephritis has been considered an autoimmune disorder of unknown origin and a complex pathophysiology, but in recent years, its pathogenesis has been unraveled. RECENT FINDINGS In individuals with unfortunate combinations of gene variants, environmental triggers such as certain drugs or viral infections allow autoimmunization against nuclear antigens, as evident by the presence of antinuclear antibodies. The expansion of autoreactive lymphocyte clones is driven by the nucleic acid component of nuclear particles from netting neutrophils and other dying cells as these activate immunity via viral nucleic acid-specific Toll-like receptors in dendritic cells and B cells. This process triggers interferon-α signaling-related antiviral immunity; hence, lupus symptoms are often indistinguishable from those of viral infections. Inside the kidney, lupus autoantibodies bind several autoantigens and trigger injury and inflammation, either in the mesangium (classes I/II), or along both sides of the glomerular basement membrane (classes III/IV/V). In addition, vascular and tubulointerstitial inflammation contribute to the immunopathology of lupus nephritis. SUMMARY Eventually, a better understanding of lupus nephritis pathogenesis will allow the identification of therapeutic targets that will prove effective not only in animal models but also in randomized clinical trials.
Collapse
|
34
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 731] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
35
|
Botte DAC, Noronha IL, Malheiros DMAC, Peixoto TV, de Mello SBV. Alpha-melanocyte stimulating hormone ameliorates disease activity in an induced murine lupus-like model. Clin Exp Immunol 2014; 177:381-90. [PMID: 24666423 DOI: 10.1111/cei.12336] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2014] [Indexed: 11/29/2022] Open
Abstract
Alpha-melanocyte stimulating hormone (α-MSH) is a neuropeptide exhibiting anti-inflammatory activity in experimental models of autoimmune diseases. However, no studies thus far have examined the effects of α-MSH on systemic lupus erythematosus (SLE). This study aimed to determine the effects of an α-MSH agonist in induced murine lupus. Here we employed female Balb/cAn mice in which lupus was induced by pristane. Groups of lupus animals were treated daily with the α-MSH analogue [Nle4, DPhe7]-α-MSH (NDP-MSH) (1·25 mg/kg) injected intraperitoneally or saline for 180 days. Normal animals comprised the control group. Arthritis incidence, plasma immunoglobulin (Ig)G isotypes, anti-nuclear antibodies (ANA) and plasma cytokines were evaluated. Renal function was assessed by proteinuria and histopathological lesion. Glomerular levels of IgG, α-smooth muscle actin (α-SMA), inducible nitric oxide synthase (iNOS), C3, CD3, melanocortin receptors (MCR)1, corticotrophin-releasing factor (CRF) and α-MSH was estimated by immunohistochemistry. When compared with normal controls, lupus animals exhibited increased arthritis, IgG levels, ANA, interleukin (IL)-6, IL-10, proteinuria and mesangial cell proliferation together with glomerular expression of α-SMA and iNOS. Glomerular expression of MCR1 was reduced in lupus animals. NDP-MSH treatment reduced arthritis scores by 70% and also diminished IgG1 and IgG2a levels and ANA incidence. In the glomerulus, NDP-MSH treatment reduced cellularity by 50% together with reducing IgG deposits, and expression levels of α-SMA, iNOS and CRF were also all decreased. Taken together, our results suggest for the first time that α-MSH treatment improves several parameters of SLE disease activity in mice, and indicate that this hormone is an interesting potential future treatment option.
Collapse
Affiliation(s)
- D A C Botte
- Rheumatology, Department of Internal Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
36
|
Pawar RD, Goilav B, Xia Y, Zhuang H, Herlitz L, Reeves WH, Putterman C. Serum autoantibodies in pristane induced lupus are regulated by neutrophil gelatinase associated lipocalin. Clin Immunol 2014; 154:49-65. [PMID: 24971701 PMCID: PMC4119527 DOI: 10.1016/j.clim.2014.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/28/2022]
Abstract
The onset of autoantibodies in systemic autoimmunity can be the result of a breakdown in tolerance at multiple checkpoints. Genetic, hormonal, and immunological factors can combine with environmental influences to accelerate the onset of disease and aggravate disease outcome. Here, we describe a novel mechanism relating to the regulatory role of Neutrophil Gelatinase Associated Lipocalin (NGAL) in modulating the levels of autoantibodies in pristane induced lupus. Following a single injection of pristane intraperitoneally, NGAL expression was induced in both the serum and spleen. Furthermore, NGAL deficient mice were more susceptible to the induction of pristane stimulated autoimmunity, and displayed higher numbers of autoantibody secreting cells and increased expression of activation induced cytidine deaminase (AID) and other inflammatory mediators in the spleen. In contrast, kidney damage was milder in NGAL deficient mice, indicating that NGAL was detrimental in autoantibody mediated kidney disease. These studies indicate that NGAL plays differential roles in different tissues in the context of lupus, and suggest a previously unrecognized role for NGAL in adaptive immunity.
Collapse
Affiliation(s)
- Rahul D Pawar
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Beatrice Goilav
- The Division of Pediatric Nephrology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yumin Xia
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haoyang Zhuang
- The Division of Rheumatology & Clinical Immunology, University of Florida, Gainesville, FL 32611, USA
| | - Leal Herlitz
- The Department of Pathology, Columbia University Medical Center, NY 10032, USA
| | - Westley H Reeves
- The Division of Rheumatology & Clinical Immunology, University of Florida, Gainesville, FL 32611, USA
| | - Chaim Putterman
- The Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
37
|
Biermann MHC, Veissi S, Maueröder C, Chaurio R, Berens C, Herrmann M, Munoz LE. The role of dead cell clearance in the etiology and pathogenesis of systemic lupus erythematosus: dendritic cells as potential targets. Expert Rev Clin Immunol 2014; 10:1151-64. [DOI: 10.1586/1744666x.2014.944162] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
38
|
Summers SA, Odobasic D, Khouri MB, Steinmetz OM, Yang Y, Holdsworth SR, Kitching AR. Endogenous interleukin (IL)-17A promotes pristane-induced systemic autoimmunity and lupus nephritis induced by pristane. Clin Exp Immunol 2014; 176:341-50. [PMID: 24528105 DOI: 10.1111/cei.12287] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2014] [Indexed: 12/20/2022] Open
Abstract
Interleukin (IL)-17A is increased both in serum and in kidney biopsies from patients with lupus nephritis, but direct evidence of pathogenicity is less well established. Administration of pristane to genetically intact mice results in the production of autoantibodies and proliferative glomerulonephritis, resembling human lupus nephritis. These studies sought to define the role of IL-17A in experimental lupus induced by pristane administration. Pristane was administered to wild-type (WT) and IL-17A(-/-) mice. Local and systemic immune responses were assessed after 6 days and 8 weeks, and autoimmunity, glomerular inflammation and renal injury were measured at 7 months. IL-17A production increased significantly 6 days after pristane injection, with innate immune cells, neutrophils (Ly6G(+)) and macrophages (F4/80(+)) being the predominant source of IL-17A. After 8 weeks, while systemic IL-17A was still readily detected in WT mice, the levels of proinflammatory cytokines, interferon (IFN)-γ and tumour necrosis factor (TNF) were diminished in the absence of endogenous IL-17A. Seven months after pristane treatment humoral autoimmunity was diminished in the absence of IL-17A, with decreased levels of immunoglobulin (Ig)G and anti-dsDNA antibodies. Renal inflammation and injury was less in the absence of IL-17A. Compared to WT mice, glomerular IgG, complement deposition, glomerular CD4(+) T cells and intrarenal expression of T helper type 1 (Th1)-associated proinflammatory mediators were decreased in IL-17A(-/-) mice. WT mice developed progressive proteinuria, but functional and histological renal injury was attenuated in the absence of IL-17A. Therefore, IL-17A is required for the full development of autoimmunity and lupus nephritis in experimental SLE, and early in the development of autoimmunity, innate immune cells produce IL-17A.
Collapse
Affiliation(s)
- S A Summers
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, VIC, Australia; Department of Nephrology, Monash Health, Melbourne, VIC, Australia
| | | | | | | | | | | | | |
Collapse
|
39
|
Chang H, Yu DS, Liu XQ, Zhang QY, Cheng N, Zhang SQ, Qu ZH. Clinical significance of TLR3 and TLR4 in peripheral blood mononuclear cells from children with Henoch-Schönlein purpura nephritis. Exp Ther Med 2014; 7:1703-1707. [PMID: 24926370 PMCID: PMC4043559 DOI: 10.3892/etm.2014.1638] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/17/2013] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to investigate the expression levels and clinical significance of Toll-like receptor (TLR) 3 and 4 in peripheral blood mononuclear cells (PBMCs) collected from children with Henoch-Schönlein purpura (HSP) nephritis. The randomized controlled trial was conducted between August 2011 and March 2013, and 105 children with a clinical diagnosis of HSP were enrolled in the study. According to the 24-h urinary protein measurements and the presence of renal damage, the 105 cases were divided into groups A, B and C as follows: Group A, children with HSP but without renal damage; group B, children with HSP nephritis but without proteinuria; group C, children with HSP nephritis and proteinuria. A total of 30 healthy children were enrolled in the normal control group (group N). The primary endpoints were the detection of TLR3 and 4 mRNA and protein expression levels in PBMCs by flow cytometry and quantitative polymerase chain reaction. The mRNA and protein expression levels of TLR4 in the PBMCs were significantly higher in groups A, B and C when compared with group N. In addition, the mRNA and protein expression levels of TLR4 in group C were much higher when compared with groups A and B. A positive correlation was identified between TLR4 protein expression and 24-h urinary protein levels in group C. The expression levels of TLR3 did not significantly differ among the groups. Protein and mRNA expression levels of TLR4 in PBMCs significantly increased and exhibited a positive correlation with urinary protein excretion. These results indicate that aberrant activation of TLR4 may be relevant to the development of HSP nephritis.
Collapse
Affiliation(s)
- Hong Chang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| | - Dong-Sheng Yu
- Department of Interventional Medicine, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| | - Xiu-Qin Liu
- Department of Pediatrics, Municipal Hospital of Qingdao, Qingdao, Shandong 266071, P.R. China
| | - Qiu-Ye Zhang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| | - Na Cheng
- Department of Pediatrics, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| | - Shou-Qing Zhang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| | - Zheng-Hai Qu
- Department of Pediatrics, The Affiliated Hospital of Qingdao University Medical College, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
40
|
Abstract
Toll-like receptors (TLRs) are pattern-recognition receptors that recognize microbial/vial-derived components that trigger innate immune response, which indicate these molecules play a role in host defense against infection. The infection often precedes numerous disorders including glomerular diseases (glomerulonephritis (GN)). It is reported that TLRs are also involved in the risk and progression of GN, and TLRs may be potential therapeutic targets for GN. To date, a number of studies have found that TLRs are involved in the pathogenesis of GN. There is a paucity of reviews in the literature discussing signaling pathways and gene expression for TLRs in GN. This review was performed to provide a relatively complete signaling pathway flowchart for TLRs to the investigators who were interested in the roles of TLRs in the pathogenesis of GN. In the past decades, some studies were also performed to explore the association of TLRs gene expression with the risk of GN. However, the role of TLRs in the pathogenesis of GN remains controversial. Here, the signal transduction pathways of TLRs and its role of gene expression in the pathogenesis of GN were reviewed.
Collapse
Affiliation(s)
- Song Mao
- Department of Nephrology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University , Nanjing , China
| | | |
Collapse
|