1
|
Tang Y, Cao Q, Liu J, Zhuang Q. Immune Landscape Variation in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Circulation Before and After Plasmapheresis by Single-Cell Transcriptome. Mediators Inflamm 2025; 2025:5531382. [PMID: 40256686 PMCID: PMC12006691 DOI: 10.1155/mi/5531382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/08/2025] [Indexed: 04/22/2025] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of autoimmune diseases characterized by inflammation and destruction of small blood vessels. AAV could be fatal if left untreated. Prompt diagnosis and treatment are crucial to protect AAV-related organs and tissue. Plasmapheresis, a therapeutic intervention aimed at removing harmful substances from the blood, devotes benefits to AAV treatment. However, the specific immune mechanism underlying its effectiveness remains unclear. In our research, we used single-cell RNA sequencing (scRNA-seq) to study the variation of peripheral blood mononuclear cells (PBMCs) before and after plasmapheresis in AAV patients. From this work, we explored a novel method for monocyte classification. In addition, flow cytometry was used to detect the relationship between the monocyte clusters and AAV activity under the new monocyte clustering method. Our scRNA-seq results revealed significant changes in monocyte clusters following treatment, which could be classified into three clusters (CD14+ monocytes, FCGR1A+ monocytes, and FCGR3A+ monocytes). In addition, our flow cytometry results showed that FCGR3A+ (CD16+) monocytes were positively correlated with AAV activity, whereas FCGR1A+ (CD16-CD64+) monocytes were negatively correlated with AAV activity. This may be related to the different biological effects of CD16 and CD64 on monocytes after interacting with the Fc region of ANCAs. In conclusion, our research sheds light on the immune landscape of AAV before and after plasmapheresis, identifying specific monocyte clusters linked to disease activity. These findings offer insights for novel monitoring methods and therapeutic targets in AAV.
Collapse
Affiliation(s)
- Youzhou Tang
- Department of Nephropathy and Rheumatology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Critical Kidney Disease Research Center, Central South University, Changsha, Hunan, China
| | - Qingtai Cao
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jishi Liu
- Department of Nephropathy and Rheumatology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan Zhuang
- Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan, China
| |
Collapse
|
2
|
Cai X, Li J, Wu M, Liu Q. Causal relationship between immune cells and the risk of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis: A Mendelian randomization study. FASEB J 2024; 38:e23821. [PMID: 39018091 DOI: 10.1096/fj.202400141r] [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: 01/18/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024]
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare inflammatory disease categorized as antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis. The majority of patients are ANCA-positive, predominantly against myeloperoxidase (MPO). Previous studies have predominantly concentrated on the association between EGPA and neutrophils, but recent research has emphasized the role of lymphocytes in the development of EGPA. The objective of our research was to examine the causal association between immune cells and MPO + ANCA EGPA. A two-sample bidirectional Mendelian randomization (MR) analysis was performed, which included 159 MPO + ANCA EGPA cases and 6688 controls and utilized Genome-Wind Associaton Studies (GWAS) summary statistics of immune traits from approximately 3757 individuals, encompassing around 22 million single nucleotide polymorphisms (SNPs). Our findings revealed that 23 immunophenotypes were associated with MPO + ANCA EGPA. Furthermore, the reverse MR analysis showed that MPO + ANCA EGPA had significant causal effects on three immunophenotypes within the Treg panel. By integrating existing research, our study unveiled the contributions of Tregs, B cells, and monocytes to the development of EGPA. Subgroup analysis specifically examined the roles of lymphocyte subtypes, cytokines, and their surface molecules in the pathogenic mechanisms of the disease. This comprehensive approach provides a novel perspective on the biological mechanisms and early intervention strategies for MPO + ANCA EGPA by focusing on immune cells.
Collapse
Affiliation(s)
- Xiaojing Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manyi Wu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Leacy EJ, Teh JW, O’Rourke AM, Brady G, Gargan S, Conlon N, Scott J, Dunne J, Phelan T, Griffin MD, Power J, Mooney A, Naughton A, Kiersey R, Gardiner M, O’Brien C, Mullan R, Flood R, Clarkson M, Townsend L, O’Shaughnessy M, Dyer AH, Moran B, Fletcher JM, Zgaga L, Little MA. Effect of Immunosuppression on the Immune Response to SARS-CoV-2 Infection and Vaccination. Int J Mol Sci 2024; 25:5239. [PMID: 38791279 PMCID: PMC11120762 DOI: 10.3390/ijms25105239] [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: 04/03/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Immunosuppressive treatment in patients with rheumatic diseases can maintain disease remission but also increase risk of infection. Their response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is frequently blunted. In this study we evaluated the effect of immunosuppression exposure on humoral and T cell immune responses to SARS-CoV-2 infection and vaccination in two distinct cohorts of patients; one during acute SARS-CoV-2 infection and 3 months later during convalescence, and another prior to SARS-CoV-2 vaccination, with follow up sampling 6 weeks after vaccination. Results were compared between rituximab-exposed (in previous 6 months), immunosuppression-exposed (in previous 3 months), and non-immunosuppressed groups. The immune cell phenotype was defined by flow cytometry and ELISA. Antigen specific T cell responses were estimated using a whole blood stimulation interferon-γ release assay. A focused post-vaccine assessment of rituximab-treated patients using high dimensional spectral cytometry was conducted. Acute SARS-CoV-2 infection was characterised by T cell lymphopenia, and a reduction in NK cells and naïve CD4 and CD8 cells, without any significant differences between immunosuppressed and non-immunosuppressed patient groups. Conversely, activated CD4 and CD8 cell counts increased in non-immunosuppressed patients with acute SARS-CoV-2 infection but this response was blunted in the presence of immunosuppression. In rituximab-treated patients, antigen-specific T cell responses were preserved in SARS-CoV-2 vaccination, but patients were unable to mount an appropriate humoral response.
Collapse
Affiliation(s)
- Emma J. Leacy
- Trinity Kidney Centre, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland (G.B.)
| | - Jia Wei Teh
- Department of Nephrology, Galway University Hospital, H91 YR71 Galway, Ireland
| | - Aoife M. O’Rourke
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.O.)
| | - Gareth Brady
- Trinity Kidney Centre, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland (G.B.)
| | - Siobhan Gargan
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Jennifer Scott
- Trinity Kidney Centre, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland (G.B.)
| | - Jean Dunne
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Thomas Phelan
- Trinity Kidney Centre, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland (G.B.)
| | - Matthew D. Griffin
- Department of Nephrology, Galway University Hospital, H91 YR71 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Julie Power
- Vasculitis Ireland Awareness, Belfast & Dublin, Ireland
| | - Aoife Mooney
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Aifric Naughton
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Rachel Kiersey
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Mary Gardiner
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Caroline O’Brien
- Department of Immunology, St. James’s Hospital, D08 NHY1 Dublin, Ireland (J.D.)
| | - Ronan Mullan
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Rheumatology, Tallaght University Hospital, D24 NR0A Dublin, Ireland
| | - Rachael Flood
- Department of Clinical Medicine, School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Rheumatology, Tallaght University Hospital, D24 NR0A Dublin, Ireland
| | - Michael Clarkson
- Department of Nephrology, Cork University Hospital, T12 DC4A Cork, Ireland
| | - Liam Townsend
- Department of Infectious Diseases, St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Michelle O’Shaughnessy
- Department of Nephrology, Galway University Hospital, H91 YR71 Galway, Ireland
- Department of Nephrology, Cork University Hospital, T12 DC4A Cork, Ireland
| | - Adam H. Dyer
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.O.)
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.M.O.)
| | - Lina Zgaga
- Department of Public Health and Primary Care, Institute of Population Health, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Mark A. Little
- Trinity Kidney Centre, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland (G.B.)
| |
Collapse
|
4
|
Smargianaki S, Elmér E, Lilliebladh S, Ohlsson S, Pettersson Å, Hellmark T, Johansson ÅCM. Disease Activity and Tendency to Relapse in ANCA-Associated Vasculitis Are Reflected in Neutrophil and Intermediate Monocyte Frequencies. J Immunol Res 2024; 2024:6648265. [PMID: 38213873 PMCID: PMC10781522 DOI: 10.1155/2024/6648265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 10/03/2023] [Accepted: 12/02/2023] [Indexed: 01/13/2024] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of autoimmune diseases with inflammation affecting small blood vessels and includes granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA). In this study, we investigated granulocyte and monocyte subsets in a large cohort of AAV patients with emphasis on disease activity and tendency to relapse. A cohort of 105 patients with GPA or MPA and 126 healthy controls (HCs) were included. Clinical and laboratory data were collected for all patients, including disease activity, tendency to relapse, and pharmacological treatment. Using flow cytometry, circulating eosinophils, basophils, neutrophils, and monocytes were assessed. The monocytes were subdivided into classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14-CD16+) monocytes. Mature (CD16high) or newly released (CD16dim) neutrophils were defined, as well as the frequency of CD177+ neutrophils. AAV patients displayed increased frequencies of intermediate monocytes, mature and newly released neutrophils, and an expanded population of CD177+ neutrophils compared to HC. MPA patients differed from GPA patients in terms of lower frequency of classical monocytes. No differences in cell frequencies regarding ANCA phenotype were observed. Paired data from 23 patients demonstrated that active disease was associated with an increased frequency of mature neutrophils and a decreased frequency of monocytes, in particular intermediate monocytes. Moreover, GPA patients with a tendency to relapse displayed an increased frequency of mature neutrophils with increased expression of CD177+. Relapsing MPA patients, on the other hand, showed decreased frequency of intermediate monocytes. Finally, rituximab treatment was associated with increased frequencies of classical and intermediate monocytes. In conclusion, AAV patients exhibit a skewing of different neutrophil and monocyte subpopulations that are associated with disease subtypes, disease activity, rituximab treatment, and propensity to relapse. These changes may contribute to the inflammatory process and could potentially be used as biomarkers for relapse prediction.
Collapse
Affiliation(s)
- Sofia Smargianaki
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University and Clinical Immunology and Transfusion Medicine, Skåne University Hospital, Lund, Sweden
| | - Evelina Elmér
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University and Clinical Immunology and Transfusion Medicine, Skåne University Hospital, Lund, Sweden
| | - Sandra Lilliebladh
- Nephrology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Sophie Ohlsson
- Nephrology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Åsa Pettersson
- Nephrology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Thomas Hellmark
- Nephrology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Åsa CM Johansson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University and Clinical Genetics and Pathology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
5
|
Daca A, Storoniak H, Dębska-Ślizień A, Kusztal MA, Krajewska M, Lisowska KA. Chemokines and Cytokines Profiles in Patients with Antineutrophil Cytoplasmic Antibodies-Associated Vasculitis: A Preliminary Study. Int J Mol Sci 2023; 24:15319. [PMID: 37894997 PMCID: PMC10607460 DOI: 10.3390/ijms242015319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The damage to small vessels in AAV and inflammatory reactions are accompanied by the release of various chemokines and cytokines. Using a flow cytometry technique, we assessed the levels of specific cytokines, namely IL-1β IL-6, IL-8, IL-10, IL12p70, and TNF, and chemokines, IFN-α, IP-10, and MIG in the serum from 9 healthy volunteers and 20 AAV patients, where 11 of the patients were not treated and evaluated at the time of diagnosis and 9 were already diagnosed and taking CY + GCS. The obtained results were then compared considering the activity of the disease, the type and titre of the ANCA antibodies, the inflammatory status, and the kidneys' condition. Amongst others, the IL-6, IL-8, IL-10, TNF, and MIG levels were much higher in the serum of AAV patients than in healthy controls, whereas the level of IL-1β was higher in healthy volunteers. Additionally, the levels of IL-6, IL-10, IP-10, and MIG negatively correlated with the eGFR level, while the level of IFN-α positively correlated with the titre of PR3-ANCA. As most of the molecules are implicated in trafficking primed neutrophils towards small vessels, looking for links between the levels of these cytokines/chemokines and the clinical symptoms of AAV may facilitate the diagnosis and predict the progression of the disease.
Collapse
Affiliation(s)
- Agnieszka Daca
- Department of Pathophysiology, Medical University of Gdańsk, 80-211 Gdansk, Poland;
| | - Hanna Storoniak
- Department of Nephrology, Transplantology, and Internal Diseases, Medical University of Gdańsk, 80-211 Gdansk, Poland; (H.S.); (A.D.-Ś.)
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology, and Internal Diseases, Medical University of Gdańsk, 80-211 Gdansk, Poland; (H.S.); (A.D.-Ś.)
| | - Mariusz Andrzej Kusztal
- Department of Nephrology and Translational Medicine, Medical University of Wrocław, 50-137 Wroclaw, Poland; (M.A.K.); (M.K.)
| | - Magdalena Krajewska
- Department of Nephrology and Translational Medicine, Medical University of Wrocław, 50-137 Wroclaw, Poland; (M.A.K.); (M.K.)
| | | |
Collapse
|
6
|
Monocyte subsets and monocyte-related chemokines in Takayasu arteritis. Sci Rep 2023; 13:2092. [PMID: 36746990 PMCID: PMC9902560 DOI: 10.1038/s41598-023-29369-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of Takayasu arteritis (TAK) is poorly understood and no previous studies have analyzed monocytes in TAK. This study evaluated monocyte subsets and monocyte-related chemokines in the peripheral blood of TAK patients and healthy controls (HC). Monocyte subsets were identified as classical (CD14+CD16-), intermediate (CD14+CD16dim), and non-classical (CD14dimCD16high) in the peripheral blood. The chemokines CCL (C-C chemokine ligand)2, CCL3, CCL4, CCL5, CCL7, CXCL (C-X-C motif ligand)10, and CX3CL (C-X3-C motif ligand)1 were measured in the sera. Thirty-two TAK patients and 30 HC were evaluated. Intermediate monocytes were higher in TAK than HC [25.0 cells ×106/L (16.7-52.0) vs. 17.2 cells ×106/L (9.2-25.3); p = 0.014]. Active disease was associated with monocytosis (p = 0.004), increased classical (p = 0.003), and intermediate (p < 0.001) subsets than HC. Prednisone reduced the percentage of non-classical monocytes (p = 0.011). TAK patients had lower CCL3 (p = 0.033) and CCL4 (p = 0.023) levels than HC, whereas CCL22 levels were higher in active TAK compared to the remission state (p = 0.008). Glucocorticoids were associated with lower CXCL10 levels (p = 0.012). In TAK, CCL4 correlated with total (Rho = 0.489; p = 0.005), classical and intermediate monocytes (Rho = 0.448; p = 0.010 and Rho = 0.412; p = 0.019). In conclusion, TAK is associated with altered counts of monocyte subsets in the peripheral blood compared to HC and CCL22 is the chemokine with the strongest association with active disease in TAK.
Collapse
|
7
|
Leacy E, Batten I, Sanelli L, McElheron M, Brady G, Little MA, Khouri H. Optimal LC-MS metabolomic profiling reveals emergent changes to monocyte metabolism in response to lipopolysaccharide. Front Immunol 2023; 14:1116760. [PMID: 37033938 PMCID: PMC10077522 DOI: 10.3389/fimmu.2023.1116760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Immunometabolism examines the links between immune cell function and metabolism. Dysregulation of immune cell metabolism is now an established feature of innate immune cell activation. Advances in liquid chromatography mass spectrometry (LC-MS) technologies have allowed discovery of unique insights into cellular metabolomics. Here we have studied and compared different sample preparation techniques and data normalisation methods described in the literature when applied to metabolomic profiling of human monocytes. Methods Primary monocytes stimulated with lipopolysaccharide (LPS) for four hours was used as a study model. Monocytes (n=24) were freshly isolated from whole blood and stimulated for four hours with lipopolysaccharide (LPS). A methanol-based extraction protocol was developed and metabolomic profiling carried out using a Hydrophilic Interaction Liquid Chromatography (HILIC) LC-MS method. Data analysis pipelines used both targeted and untargeted approaches, and over 40 different data normalisation techniques to account for technical and biological variation were examined. Cytokine levels in supernatants were measured by ELISA. Results This method provided broad coverage of the monocyte metabolome. The most efficient and consistent normalisation method was measurement of residual protein in the metabolite fraction, which was further validated and optimised using a commercial kit. Alterations to the monocyte metabolome in response to LPS can be detected as early as four hours post stimulation. Broad and profound changes in monocyte metabolism were seen, in line with increased cytokine production. Elevated levels of amino acids and Krebs cycle metabolites were noted and decreases in aspartate and β-alanine are also reported for the first time. In the untargeted analysis, 154 metabolite entities were significantly altered compared to unstimulated cells. Pathway analysis revealed the most prominent changes occurred to (phospho-) inositol metabolism, glycolysis, and the pentose phosphate pathway. Discussion These data report the emergent changes to monocyte metabolism in response to LPS, in line with reports from later time points. A number of these metabolites are reported to alter inflammatory gene expression, which may facilitate the increases in cytokine production. Further validation is needed to confirm the link between metabolic activation and upregulation of inflammatory responses.
Collapse
Affiliation(s)
- Emma Leacy
- Trinity Translational Medicine Institute, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Emma Leacy, ; Mark A. Little,
| | - Isabella Batten
- Trinity Translational Medicine Institute, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Laetitia Sanelli
- Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Matthew McElheron
- Trinity Translational Medicine Institute, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Gareth Brady
- Trinity Translational Medicine Institute, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Mark A. Little
- Trinity Translational Medicine Institute, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Health Kidney Centre, Tallaght University Hospital, Dublin, Ireland
- *Correspondence: Emma Leacy, ; Mark A. Little,
| | - Hania Khouri
- Agilent Technologies, Stockpoty, England, United Kingdom
| |
Collapse
|
8
|
Bai X, Xu PC, Chen T, Zhang HM, Wu SJ, Yang X, Gao S, Jia JY, Jiang JQ, Yan TK. The potential pathogenic roles of S100A8/A9 and S100A12 in patients with MPO-ANCA-positive vasculitis. BMC Immunol 2022; 23:42. [PMID: 36088289 PMCID: PMC9464401 DOI: 10.1186/s12865-022-00513-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/22/2022] [Indexed: 11/19/2022] Open
Abstract
Background The significance of S100A8/A9 and S100A12 in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) has not been clarified. This study was dedicated to exploring the potential pathogenic roles of S100A8/A9 and S100A12 in patients with myeloperoxidase (MPO)-ANCA-positive vasculitis. Methods Serum and urine concentrations of S100A8/A9 and S100A12 of forty-two AAV patients were evaluated. The influence of S100A8/A9 and S100A12 on the chemotaxis, the apoptosis, the release of IL-1β, the complement activation, the respiratory burst, as well as the neutrophil extracellular traps (NETs) formation of MPO-ANCA-activated neutrophils was investigated. Results The serum and urine S100A8/A9 and S100A12 of active MPO-AAV significantly increased (compared with inactive AAV and healthy controls, p < 0.001) and were correlated with the severity of the disease. In vitro study showed that S100A8/A9 and S100A12 activated the p38 MAPK/NF-κB p65 pathway, increased the chemotaxis index (CI) and the release of IL-1β, extended the life span, and enhanced the complement activation ability of MPO-ANCA-activated neutrophils. The Blockade of TLR4 and RAGE inhibited the effects of S100A8/A9 and S100A12. All above-mentioned effects of S100A8/A9 and S100A12 were ROS-independent because neither S100A8/A9 nor S100A12 enhanced the ROS formation and NETs formation of MPO-ANCA-activated neutrophils. Conclusion S100A8/A9 and S100A12 serve as markers for assessing the disease severity, and they may also play a role in MPO-AAV pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-022-00513-4.
Collapse
|
9
|
Tang J, Liao Z, Luo L, Deng S, Jiang Y, Wang F, Hu X, Yin H, Gong G, Feng J, Li X. CX3CL1-induced CD16+ monocytes extravasation in myeloperoxidase-ANCA-associated vasculitis correlates with renal damage. Front Immunol 2022; 13:929244. [PMID: 36059489 PMCID: PMC9437287 DOI: 10.3389/fimmu.2022.929244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background Monocytes are involved in the pathogenesis of ANCA-associated vasculitis (AAV). Monocyte/macrophages are the dominant infiltrating cells in the glomeruli of patients with myeloperoxidase-AAV (MPO-AAV). However, how human monocyte subsets extravasate to the kidney in MPO-AAV with renal damage is unclear. Methods 30 MPO-AAV patients with renal damage and 22 healthy controls were enrolled in this study. Monocyte subsets and monocyte-related chemokines in the blood and kidneys of MPO-AAV patients were detected. The chemoattractant activity of the CX3CL1-CX3CR1 axis on CD16+ monocytes was observed. The effect of MPO-ANCA on the migration of CD16+ monocytes to human glomerular endothelial cells (HGECs) was detected by flow cytometry and transwell migration assay. Results Compared with controls, CD16+ monocytes were significantly decreased in the blood and increased in the glomeruli of MPO-AAV patients with renal damage. The level of CX3CL1, but not CCL2, was significantly increased in the plasma of MPO-AAV patients. CX3CL1 co-localized with glomerular endothelial cells in MPO-AAV patients with renal damage. Moreover, we initially found that MPO-ANCA promotes an increase of the chemokine CX3CL1 on HGECs, imposing recruitment on CD16+ monocytes. Finally, the percentage of CD16+ monocytes in the blood was found to be positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with urinary protein creatinine ratio in MPO-AAV patients with renal damage. Furthermore, the urinary protein creatinine ratio was positively correlated with the infiltrating of CD14+ and CD16+ cells in the kidneys. Conclusion Enhanced extravasation of CD16+ monocytes to the kidney via the CX3CL1-CX3CR1 axis may be involved in renal damage in MPO-AAV.
Collapse
Affiliation(s)
- Jiale Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhonghua Liao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Liying Luo
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuanglinzi Deng
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Jiang
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Fangyuan Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinyue Hu
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Hongling Yin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Juntao Feng
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaozhao Li
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaozhao Li,
| |
Collapse
|
10
|
Clinical Significance of Antineutrophil Cytoplasmic Antibody Positivity in Patients Infected with SARS-CoV-2. J Clin Med 2022; 11:jcm11144152. [PMID: 35887916 PMCID: PMC9322989 DOI: 10.3390/jcm11144152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives: To investigate the rate of antineutrophil cytoplasmic antibody (ANCA) positivity and its clinical significance in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Methods: This study included 178 patients infected with SARS-CoV-2 who were enrolled in a cohort at a single centre. Myeloperoxidase (MPO)-ANCA and proteinase 3 (PR3)-ANCA levels in stored blood sera were measured using immunoassay kits. Mortality, mechanical ventilator care, and severe infection were assessed as three poor outcomes. The 2022 American College of Rheumatology and the European Alliance of Associations for Rheumatology (ACR/EULAR) classification criteria for the three subtypes of AAV were applied only to patients who had MPO-ANCA or PR3-ANCA among study subjects. Results: The detection rate of ANCA positivity was 18.5%. MPO-ANCA and PR3-ANCA were found in 22 (12.4%) and 14 (7.9%) patients, respectively. However, neither MPO-ANCA nor PR3-ANCA affected the three poor outcomes. According to the new criteria, 12 (6.7%) and 21 (11.8%) patients were classified as having granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA), respectively. Conclusions: SARS-CoV-2 infection may increase the rate of ANCA positivity. Although it might not affect poor outcomes, it might contribute to the classification of GPA and MPA despite uncertain clinical significance.
Collapse
|
11
|
Xu Y, Chen Y, Zhang X, Ma J, Liu Y, Cui L, Wang F. Glycolysis in Innate Immune Cells Contributes to Autoimmunity. Front Immunol 2022; 13:920029. [PMID: 35844594 PMCID: PMC9284233 DOI: 10.3389/fimmu.2022.920029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases (AIDs) refer to connective tissue inflammation caused by aberrant autoantibodies resulting from dysfunctional immune surveillance. Most of the current treatments for AIDs use non-selective immunosuppressive agents. Although these therapies successfully control the disease process, patients experience significant side effects, particularly an increased risk of infection. There is a great need to study the pathogenesis of AIDs to facilitate the development of selective inhibitors for inflammatory signaling to overcome the limitations of traditional therapies. Immune cells alter their predominant metabolic profile from mitochondrial respiration to glycolysis in AIDs. This metabolic reprogramming, known to occur in adaptive immune cells, i.e., B and T lymphocytes, is critical to the pathogenesis of connective tissue inflammation. At the cellular level, this metabolic switch involves multiple signaling molecules, including serine-threonine protein kinase, mammalian target of rapamycin, and phosphoinositide 3-kinase. Although glycolysis is less efficient than mitochondrial respiration in terms of ATP production, immune cells can promote disease progression by enhancing glycolysis to satisfy cellular functions. Recent studies have shown that active glycolytic metabolism may also account for the cellular physiology of innate immune cells in AIDs. However, the mechanism by which glycolysis affects innate immunity and participates in the pathogenesis of AIDs remains to be elucidated. Therefore, we reviewed the molecular mechanisms, including key enzymes, signaling pathways, and inflammatory factors, that could explain the relationship between glycolysis and the pro-inflammatory phenotype of innate immune cells such as neutrophils, macrophages, and dendritic cells. Additionally, we summarize the impact of glycolysis on the pathophysiological processes of AIDs, including systemic lupus erythematosus, rheumatoid arthritis, vasculitis, and ankylosing spondylitis, and discuss potential therapeutic targets. The discovery that immune cell metabolism characterized by glycolysis may regulate inflammation broadens the avenues for treating AIDs by modulating immune cell metabolism.
Collapse
Affiliation(s)
- Yue Xu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yongkang Chen
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Fang Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Koike H, Furukawa S, Mouri N, Fukami Y, Iijima M, Katsuno M. Early ultrastructural lesions of anti-neutrophil cytoplasmic antibody- versus complement-associated vasculitis. Neuropathology 2022; 42:420-429. [PMID: 35707831 DOI: 10.1111/neup.12821] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/24/2022] [Accepted: 05/01/2022] [Indexed: 12/11/2022]
Abstract
This study aims to describe electron microscopic findings of vasculitis associated with anti-neutrophil cytoplasmic antibody (ANCA) and complement. Sural nerve biopsy specimens were obtained from 10 patients with microscopic polyangiitis (MPA), a representative ANCA-associated vasculitis, and six patients with nonsystemic vasculitic neuropathy (NSVN), who were negative for ANCA but positive for complement deposition. In patients with MPA, attachment of neutrophils to epineurial vascular endothelial cells, migration of neutrophils to the extravascular space via the penetration of the endothelial layer, and release of neutrophil components to the extracellular space were observed. Such neutrophil-associated lesions were not observed in patients with NSVN. Nonetheless, morphological changes in epineurial vascular endothelial cells, such as increases in cytoplasmic organelles and cytoplasmic protrusions into the vascular lumen, were observed in patients with NSVN. Since these findings were observed where light microscopy-based findings suggestive of vasculitis (e.g., the disruption of vascular structures and fibrinoid necrosis) were absent, they were considered early lesions that preceded the formation of the so-called necrotizing vasculitis. In conclusion, this study enabled the visualization of distinctive early ultrastructural lesions associated with ANCA and complement. Further studies are needed to elucidate the molecular basis of the induction of these fine structural changes, which will contribute to the development of targeted therapies based on specific mechanisms of vasculitis.
Collapse
Affiliation(s)
- Haruki Koike
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Soma Furukawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naohiro Mouri
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Fukami
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Iijima
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
Massicotte-Azarniouch D, Herrera CA, Jennette JC, Falk RJ, Free ME. Mechanisms of vascular damage in ANCA vasculitis. Semin Immunopathol 2022; 44:325-345. [PMID: 35254509 PMCID: PMC9064952 DOI: 10.1007/s00281-022-00920-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
The discovery of anti-neutrophil cytoplasmic antibodies (ANCA) and their antigenic targets, myeloperoxidase (MPO) and proteinase 3 (PR3), has led to further understanding as to the pathophysiologic processes that underlie vascular and tissue damage in ANCA vasculitis. ANCA trigger neutrophil activation leading to vascular damage in ANCA vasculitis. However, decades of study have determined that neutrophil activation alone is not sufficient to cause disease. Inflammatory stimuli are drivers of ANCA autoantigen expression and ANCA production. Certain infections or bacterial peptides may be crucial players in the initial steps of ANCA immunopathogenesis. Genetic and epigenetic alterations of gene encoding for MPO and PR3 provide additional disturbances to the immune homeostasis which provide a substrate for pathogenic ANCA formation from an adaptive immune system predisposed to autoreactivity. Promoted by inflammatory cytokines, ANCA binding leads to neutrophil activation, a process characterized by conformational changes, production and release of cytotoxic substances, and alternative complement pathway activation, thus creating an intense inflammatory milieu. This cascade of events perpetuates a vicious cycle of further inflammatory cell recruitment and activation, culminating in tissue necrosis. Our understanding of the pathogenic process in ANCA vasculitis paves the way for the development of therapies targeting crucial steps in this process. The greater appreciation of the role for complement, monocytes, and the adaptive immune system has already led to novel complement blockers and is poised to lead to further innovations which will allow for tailored antigen- or cell-specific immunotherapy targeting the autoimmune process without exposure to undue risks or toxicities.
Collapse
Affiliation(s)
- David Massicotte-Azarniouch
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Carolina A Herrera
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Charles Jennette
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ronald J Falk
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Meghan E Free
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Tampe D, Hakroush S, Biggemann L, Winkler MS, Tampe B. Kinetics of human leukocyte antigen receptor HLA-DR + monocytes and T lymphocytes during remission induction therapy in ANCA-associated vasculitis. J Nephrol 2022; 35:1283-1287. [PMID: 35445945 PMCID: PMC9107415 DOI: 10.1007/s40620-022-01330-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/03/2022] [Indexed: 01/07/2023]
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis is characterized by small vessel inflammation and the presence of autoantibodies against cytoplasmic proteases, most often proteinase-3 and myeloperoxidase. Peripheral blood monocytes are an important source of local macrophage accumulation within parenchymal organs, as evidenced by their presence in early lesions in ANCA-associated glomerulonephritis. Major histocompatibility complex (MHC) II cell surface receptor human leukocyte antigen receptor (HLA-DR) allows antigen presentation to T cells and is crucial for the initiation of an immune response. We herein report HLA-DR abundance in AAV and the kinetics of HLA-DR+ monocytes and T lymphocytes during remission induction therapy in AAV. Life-threatening AAV with pulmonary hemorrhage and renal involvement was associated with the presence of HLA-DR in a considerable population of peripheral blood monocytes and T lymphocytes, and relapsing disease manifested despite persistent B cell depletion after remission induction with rituximab. Moreover, remission induction in AAV with steroids, plasma exchange and intravenous cyclophosphamide, and improvement of clinical symptoms were associated with a decrease in HLA-DR+ differing between monocytes and T lymphocytes. Particularly, persistent suppression of HLA-DR+ monocytes was observed during remission induction, while an initial decrease in HLA-DR+ T lymphocytes was followed by recovery of this population during the further course. Detailed insights into HLA-DR kinetics could pave the way towards an increased understanding of immunopathology and identify patients that could mostly benefit from distinct remission induction regimens.
Collapse
Affiliation(s)
- Désirée Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Samy Hakroush
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Lorenz Biggemann
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Sebastian Winkler
- Department of Anesthesiology, Emergency and Intensive Care Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
15
|
d’Alessandro M, Conticini E, Bergantini L, Cameli P, Cantarini L, Frediani B, Bargagli E. Neutrophil Extracellular Traps in ANCA-Associated Vasculitis and Interstitial Lung Disease: A Scoping Review. Life (Basel) 2022; 12:317. [PMID: 35207604 PMCID: PMC8877891 DOI: 10.3390/life12020317] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Deregulated neutrophil extracellular traps (NETs) formation is implicated in various diseases, including ANCA-associated vasculitis and pulmonary fibrosis (PF). Lung involvement is frequent in AAV, and interstitial lung diseases (ILDs) are strongly related to MPO-ANCA positivity and mainly reported in microscopic polyangiitis. The association between AAV and ILD is a strong indicator of poor prognosis and limited survival. Neutrophils, ANCA and NET interplay in PF development in AAV. This study aimed to review the literature concerning the implications of NET in lung fibrogenesis specifically focused on AAV associated with ILD, and the potential of NET as a theranostic marker. METHODS Through scoping review methodology, we used a descriptive thematic analysis to understand the pathogenic role of NETs in patients with AAV and pulmonary fibrosis and their further role as a theranostic marker of this disease. RESULTS The implications of NET in the pathogenesis of AAV and ILD, as well as an association between these two diseases, have been identified, but the underlying pathophysiological mechanisms are still unknown. The pharmacological or genetic inhibition of NET release reduces disease severity in multiple inflammatory disease models, indicating that NETs are potential therapeutic targets. In this regard, despite the lack of clinical data, we may hypothesise that an optimal management of AAV-ILD patients would require not only B-cells targeted therapy, but also NETs inhibition. CONCLUSION Preliminary findings seem to display a lack of efficacy of traditional immunosuppressants, such as Rituximab, in this subset of patients, while to date no patients suffering from a definite ILD have been enrolled in clinical trials. Further insights would be provided by their employment, as a combination treatment, in common clinical practice. Although we can imagine that the inhibition of NETs in patients with AAV-ILD could reduce severity and mortality, we still lack the scientific basis that could improve our understanding of the disease from a molecular point of view.
Collapse
Affiliation(s)
- Miriana d’Alessandro
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, University of Siena, 53100 Siena, Italy; (M.d.); (L.B.); (P.C.); (E.B.)
| | - Edoardo Conticini
- Rheumatology Unit, Department of Medicine, Surgery & Neurosciences, University of Siena, 53100 Siena, Italy; (L.C.); (B.F.)
| | - Laura Bergantini
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, University of Siena, 53100 Siena, Italy; (M.d.); (L.B.); (P.C.); (E.B.)
| | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, University of Siena, 53100 Siena, Italy; (M.d.); (L.B.); (P.C.); (E.B.)
| | - Luca Cantarini
- Rheumatology Unit, Department of Medicine, Surgery & Neurosciences, University of Siena, 53100 Siena, Italy; (L.C.); (B.F.)
| | - Bruno Frediani
- Rheumatology Unit, Department of Medicine, Surgery & Neurosciences, University of Siena, 53100 Siena, Italy; (L.C.); (B.F.)
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, University of Siena, 53100 Siena, Italy; (M.d.); (L.B.); (P.C.); (E.B.)
| |
Collapse
|
16
|
Box CD, Cronin O, Hauser B. The Impact of High Dose Glucocorticoids on Bone Health and Fracture Risk in Systemic Vasculitides. Front Endocrinol (Lausanne) 2022; 13:806361. [PMID: 35250864 PMCID: PMC8889574 DOI: 10.3389/fendo.2022.806361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 12/29/2022] Open
Abstract
Systemic vasculitides are a range of conditions characterized by inflammation of blood vessels which may manifest as single organ or life-threatening multisystem disease. The treatment of systemic vasculitis varies depending on the specific disease but historically has involved initial treatment with high dose glucocorticoids alone or in conjunction with other immunosuppressive agents. Prolonged glucocorticoid treatment is frequently required as maintenance treatment. Patients with small and large vessel vasculitis are at increased risk of fracture. Osteoporosis may occur due to intrinsic factors such as chronic inflammation, impaired renal function and to a large extent due to pharmacological therapy with high dose glucocorticoid or combination treatments. This review will outline the known mechanism of bone loss in vasculitis and will summarize factors attributing to fracture risk in different types of vasculitis. Osteoporosis treatment with specific consideration for patients with vasculitis will be discussed. The use of glucocorticoid sparing immunosuppressive agents in the treatment of systemic vasculitis is a significant area of ongoing research. Adjunctive treatments are used to reduce cumulative doses of glucocorticoids and therefore may significantly decrease the associated fracture risk in patients with vasculitis. Lastly, we will highlight the many unknowns in the relation between systemic vasculitis, its treatment and bone health and will outline key research priorities for this field.
Collapse
Affiliation(s)
| | - Owen Cronin
- Department of Rheumatology, Bon Secours Hospital Cork, Cork, Ireland
- School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Barbara Hauser
- Rheumatic Disease Unit, Western General Hospital, Edinburgh, United Kingdom
- Rheumatology and Bone Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Barbara Hauser,
| |
Collapse
|
17
|
Chalayer E, Gramont B, Zekre F, Goguyer-Deschaumes R, Waeckel L, Grange L, Paul S, Chung AW, Killian M. Fc receptors gone wrong: A comprehensive review of their roles in autoimmune and inflammatory diseases. Autoimmun Rev 2021; 21:103016. [PMID: 34915182 DOI: 10.1016/j.autrev.2021.103016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022]
Abstract
Systemic autoimmune and inflammatory diseases have a complex and only partially known pathophysiology with various abnormalities involving all the components of the immune system. Among these components, antibodies, and especially autoantibodies are key elements contributing to autoimmunity. The interaction of antibody fragment crystallisable (Fc) and several distinct receptors, namely Fc receptors (FcRs), have gained much attention during the recent years, with possible major therapeutic perspectives for the future. The aim of this review is to comprehensively describe the known roles for FcRs (activating and inhibitory FcγRs, neonatal FcR [FcRn], FcαRI, FcεRs, Ro52/tripartite motif containing 21 [Ro52/TRIM21], FcδR, and the novel Fc receptor-like [FcRL] family) in systemic autoimmune and inflammatory disorders, namely rheumatoid arthritis, Sjögren's syndrome, systemic lupus erythematosus, systemic sclerosis, idiopathic inflammatory myopathies, mixed connective tissue disease, Crohn's disease, ulcerative colitis, immunoglobulin (Ig) A vasculitis, Behçet's disease, Kawasaki disease, IgG4-related disease, immune thrombocytopenia, autoimmune hemolytic anemia, antiphospholipid syndrome and heparin-induced thrombocytopenia.
Collapse
Affiliation(s)
- Emilie Chalayer
- Department of Hematology and Cell Therapy, Institut de Cancérologie Lucien Neuwirth, Saint-Etienne, France; INSERM U1059-Sainbiose, dysfonction vasculaire et hémostase, Université de Lyon, Saint-Etienne, France
| | - Baptiste Gramont
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Franck Zekre
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Pediatrics, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Roman Goguyer-Deschaumes
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Lucile Grange
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Amy W Chung
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Martin Killian
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France.
| |
Collapse
|
18
|
Morissette F, Mongeau-Pérusse V, Rizkallah E, Thébault P, Lepage S, Brissette S, Bruneau J, Dubreucq S, Stip E, Cailhier JF, Jutras-Aswad D. Exploring cannabidiol effects on inflammatory markers in individuals with cocaine use disorder: a randomized controlled trial. Neuropsychopharmacology 2021; 46:2101-2111. [PMID: 34331010 PMCID: PMC8505631 DOI: 10.1038/s41386-021-01098-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/29/2023]
Abstract
Cocaine use disorder (CUD) is a major public health issue associated with physical, social, and psychological problems. Excessive and repeated cocaine use induces oxidative stress leading to a systemic inflammatory response. Cannabidiol (CBD) has gained substantial interest for its anti-inflammatory properties, safety, and tolerability profile. However, CBD anti-inflammatory properties have yet to be confirmed in humans. This exploratory study is based on a single-site randomized controlled trial that enrolled participants with CUD between 18 and 65 years, randomized (1:1) to daily receive either CBD (800 mg) or placebo for 92 days. The trial was divided into a 10-day detoxification (phase I) followed by a 12-week outpatient follow-up (phase II). Blood samples were collected from 48 participants at baseline, day 8, week 4, and week 12 and were analyzed to determine monocytes and lymphocytes phenotypes, and concentrations of various inflammatory markers such as cytokines. We used generalized estimating equations to detect group differences. Participants treated with CBD had lower levels of interleukin-6 (p = 0.017), vascular endothelial growth factor (p = 0.032), intermediate monocytes CD14+CD16+ (p = 0.024), and natural killer CD56negCD16hi (p = 0.000) compared with participants receiving placebo. CD25+CD4+T cells were higher in the CBD group (p = 0.007). No significant group difference was observed for B lymphocytes. This study suggests that CBD may exert anti-inflammatory effects in individuals with CUD.
Collapse
Affiliation(s)
- Florence Morissette
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Violaine Mongeau-Pérusse
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Elie Rizkallah
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Paméla Thébault
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada
| | - Stéphanie Lepage
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada
| | - Suzanne Brissette
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Family and Emergency Medicine, Université de Montréal, Montreal, QC Canada
| | - Julie Bruneau
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Family and Emergency Medicine, Université de Montréal, Montreal, QC Canada
| | - Simon Dubreucq
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada
| | - Emmanuel Stip
- grid.14848.310000 0001 2292 3357Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC Canada ,grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,grid.43519.3a0000 0001 2193 6666Department of Psychiatry and Behavioral Science, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Jean-François Cailhier
- grid.410559.c0000 0001 0743 2111Research Centre of Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC Canada ,Montreal Cancer Institute, Montreal, QC Canada ,grid.14848.310000 0001 2292 3357Division of Nephrology, Department of Medicine, Université de Montréal, Montreal, QC Canada
| | - Didier Jutras-Aswad
- Faculty of Medicine, Department of Psychiatry and Addictology, Université de Montréal, Montreal, QC, Canada. .,Research Centre of Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada. .,University Institute on Addictions, Montreal, QC, Canada.
| |
Collapse
|
19
|
Distribution of monocytes subpopulations in the peripheral blood from patients with Behçet's disease - Impact of disease status and colchicine use. Clin Immunol 2021; 231:108854. [PMID: 34530137 DOI: 10.1016/j.clim.2021.108854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022]
Abstract
The innate immune response has a predominant role in Behçet's disease (BD) pathogenesis, but few studies have assessed monocytes in BD. This study aims to evaluate the profile of monocytes subsets in the peripheral blood of BD patients and healthy controls (HC). Monocytes subsets were identified as classical (CD14+CD16-), intermediate (CD14+CD16dim), and non-classical (CD14dimCD16high) subsets. Patients with BD presented a lower number of total monocytes (p = 0.020) and a lower number (p < 0.0001) of circulating classical monocytes than HC. In contrast, the number of intermediate monocytes was higher in BD patients than HC (p < 0.0001). In BD patients, no associations were observed with the severity of clinical manifestations or therapy. Colchicine was associated with a higher number of non-classical monocytes (p = 0.035). In conclusion, BD patients present an altered distribution of monocytes subsets with a reduction of classical and an increase of intermediate subsets.
Collapse
|
20
|
Pyo JY, Lee LE, Ahn SS, Song JJ, Park YB, Lee SW. Efficacy of tacrolimus as maintenance therapy after cyclophosphamide for treating antineutrophil cytoplasmic antibody-associated vasculitis. Medicine (Baltimore) 2021; 100:e26956. [PMID: 34449460 PMCID: PMC8389966 DOI: 10.1097/md.0000000000026956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/29/2021] [Indexed: 01/04/2023] Open
Abstract
Azathioprine (AZA), methotrexate, or rituximab is used for the maintenance therapy of antineutrophil cytoplasmic antibody-associated vasculitis (AAV). Although the efficacy of tacrolimus (TAC) in various autoimmune diseases has been demonstrated, there have been few reports on the efficacy of TAC in AAV. We investigated the efficacy of TAC as maintenance therapy for AAV and compared its efficacy with that of AZA.We retrospectively analyzed the medical records of 81 patients with AAV who received cyclophosphamide as induction therapy and AZA or TAC as maintenance therapy. All-cause death, relapse, and progression to end-stage renal disease (ESRD) were analyzed.Among 81 patients with AAV, 69 patients received AZA alone, 6 patients received TAC alone, and 6 patients received TAC after AZA for maintenance therapy. Overall, 11 patients (13.6%) died, 30 patients (37.0%) experienced relapse, and 16 patients (19.8%) progressed to ESRD during a median of 33.8 months. No significant differences were observed in cumulative patients', relapse-free, and ESRD-free survival rates between patients administered AZA alone and TAC alone. There were no significant differences in the cumulative patients' and relapse-free survival rate between patients who received AZA alone and TAC after AZA. However, the cumulative ESRD-free survival rate was lower in patients who received TAC after AZA than in those who received AZA alone (P = .027).Patients who received TAC as maintenance therapy showed a higher incidence of ESRD than those who received AZA; however, this might be attributed to the lack of efficacy of AZA rather than the low ESRD prevention effect of TAC.
Collapse
Affiliation(s)
- Jung Yoon Pyo
- Division of Rheumatology, Department of Internal Medicine
| | - Lucy Eunju Lee
- Division of Rheumatology, Department of Internal Medicine
| | - Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
Vegting Y, Vogt L, Anders HJ, de Winther MPJ, Bemelman FJ, Hilhorst ML. Monocytes and macrophages in ANCA-associated vasculitis. Autoimmun Rev 2021; 20:102911. [PMID: 34298153 DOI: 10.1016/j.autrev.2021.102911] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 06/05/2021] [Indexed: 12/15/2022]
Abstract
Anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides (AAV) are characterized by inflammation of small-to-medium-sized blood vessels and the presence of autoantibodies against cytoplasmic proteases sited in neutrophils and monocytes. Increasing evidence indicates a substantial role of monocytes and macrophages in the pathogenesis of AAV. Activated monocytes and macrophages contribute to necroinflammation in peripheral vasculitic lesions as well as to central and peripheral mechanisms of autoimmunity. The intermediate monocyte subset (CD14++CD16+) is increased and monocytes show elevated expression of CD14, Toll-like receptor 2/4, MHCII and integrins, likely reflecting activation and increased monocyte extravasation. Monocytes differentiate locally predominantly into alternatively activated (M2) macrophages, which are known for cell-clearance and phagocytosis, but may ultimately lead to fibrosis. Phagocytotic function of macrophages can be impaired by surface expression of cytoplasmic proteases on apoptotic neutrophils and causes release of inflammatory cytokines and immunogenic contents, presumably resulting in a vicious circle of increased neutrophil, T and B cell activation and consequent ANCA production. Considering their crucial role in initiating necroinflammation as well as fibrogenesis, monocytes and macrophages may represent a logic first-line target for new treatment options in AAV.
Collapse
Affiliation(s)
- Yosta Vegting
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hans-Joachim Anders
- Department of Internal Medicine IV, Division of Nephrology, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike J Bemelman
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marc L Hilhorst
- Department of Internal Medicine, Section of Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Müller-Deile J, Jaremenko C, Haller H, Schiffer M, Haubitz M, Christiansen S, Falk C, Schiffer L. Chemokine/Cytokine Levels Correlate with Organ Involvement in PR3-ANCA-Associated Vasculitis. J Clin Med 2021; 10:jcm10122715. [PMID: 34205404 PMCID: PMC8234887 DOI: 10.3390/jcm10122715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/07/2021] [Accepted: 06/17/2021] [Indexed: 02/02/2023] Open
Abstract
Background: ANCA-associated vasculitis (AAV) is a rare small vessel disease characterized by multi-organ involvement. Biomarkers that can measure specific organ involvement are missing. Here, we ask whether certain circulating cytokines and chemokines correlate with renal involvement and if distinct cytokine/chemokine patterns can differentiate between renal, ear/nose/throat, joints, and lung involvement of AAV. Methods: Thirty-two sets of Birmingham vasculitis activity score (BVAS), PR3-ANCA titers, laboratory marker, and different cytokines were obtained from 17 different patients with AAV. BVAS, PR3-ANCA titers, laboratory marker, and cytokine concentrations were correlated to different organ involvements in active AAV. Results: Among patients with active PR3-AAV (BVAS > 0) and kidney involvement we found significant higher concentrations of chemokine ligand (CCL)-1, interleukin (IL)-6, IL21, IL23, IL-28A, IL33, monocyte chemoattractant protein 2 (MCP2), stem cell factor (SCF), thymic stromal lymphopoietin (TSLP), and thrombopoietin (TPO) compared to patients without PR3-ANCA-associated glomerulonephritis. Patients with ear, nose, and throat involvement expressed higher concentrations of MCP2 and of the (C-X-C motif) ligand-12 (CXCL-12) compared to patients with active AAV and no involvement of these organs. Conclusion: We identified distinct cytokine patterns for renal manifestation and for ear, nose and throat involvement of PR3-AAV. Distinct plasma cytokines might be used as non-invasive biomarkers of organ involvement in AAV.
Collapse
Affiliation(s)
- Janina Müller-Deile
- Department of Nephrology and Hypertension, Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany;
- Correspondence:
| | - Christian Jaremenko
- Institute for Nanotechnology and Correlative Microscopy eV, INAM, 91301 Forchheim, Germany; (C.J.); (S.C.)
- Institute of Optics, Information and Photonics, Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Hermann Haller
- Department of Nephrology, Hannover Medical School, 30625 Hannover, Germany;
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander-University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Marion Haubitz
- Department of Nephrology and Hypertension, Center for Internal Medicine and Medical Clinic III, Klinikum Fulda, 36043 Fulda, Germany;
| | - Silke Christiansen
- Institute for Nanotechnology and Correlative Microscopy eV, INAM, 91301 Forchheim, Germany; (C.J.); (S.C.)
| | - Christine Falk
- Institute of Transplant Immunology, Hannover Medical School, 30625 Hannover, Germany;
| | - Lena Schiffer
- Department of Pediatric Nephrology, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
23
|
Müller A, Krause B, Kerstein-Stähle A, Comdühr S, Klapa S, Ullrich S, Holl-Ulrich K, Lamprecht P. Granulomatous Inflammation in ANCA-Associated Vasculitis. Int J Mol Sci 2021; 22:ijms22126474. [PMID: 34204207 PMCID: PMC8234846 DOI: 10.3390/ijms22126474] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022] Open
Abstract
ANCA-associated vasculitis (AAV) comprises granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). While systemic vasculitis is a hallmark of all AAV, GPA is characterized by extravascular granulomatous inflammation, preferentially affecting the respiratory tract. The mechanisms underlying the emergence of neutrophilic microabscesses; the appearance of multinucleated giant cells; and subsequent granuloma formation, finally leading to scarred or destroyed tissue in GPA, are still incompletely understood. This review summarizes findings describing the presence and function of molecules and cells contributing to granulomatous inflammation in the respiratory tract and to renal inflammation observed in GPA. In addition, factors affecting or promoting the development of granulomatous inflammation such as microbial infections, the nasal microbiome, and the release of damage-associated molecular patterns (DAMP) are discussed. Further, on the basis of numerous results, we argue that, in situ, various ways of exposure linked with a high number of infiltrating proteinase 3 (PR3)- and myeloperoxidase (MPO)-expressing leukocytes lower the threshold for the presentation of an altered PR3 and possibly also of MPO, provoking the local development of ANCA autoimmune responses, aided by the formation of ectopic lymphoid structures. Although extravascular granulomatous inflammation is unique to GPA, similar molecular and cellular patterns can be found in both the respiratory tract and kidney tissue of GPA and MPA patients; for example, the antimicrobial peptide LL37, CD163+ macrophages, or regulatory T cells. Therefore, we postulate that granulomatous inflammation in GPA or PR3-AAV is intertwined with autoimmune and destructive mechanisms also seen at other sites.
Collapse
Affiliation(s)
- Antje Müller
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Correspondence: ; Tel.: +49-451-5005-0867
| | - Bettina Krause
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Institute of Anatomy & Experimental Morphology, University Hospital Hamburg-Eppendorf, University of Hamburg, 20251 Hamburg, Germany;
| | - Anja Kerstein-Stähle
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| | - Sara Comdühr
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| | - Sebastian Klapa
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Institute of Experimental Medicine c/o German Naval Medical Institute, Carl-Albrechts University of Kiel, 24119 Kronshagen, Germany
| | - Sebastian Ullrich
- Institute of Anatomy & Experimental Morphology, University Hospital Hamburg-Eppendorf, University of Hamburg, 20251 Hamburg, Germany;
- Municipal Hospital Kiel, 24116 Kiel, Germany
| | | | - Peter Lamprecht
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| |
Collapse
|
24
|
Page TH, Chiappo D, Brunini F, Garnica J, Blackburn J, Dudhiya F, Prendecki M, McAdoo SP, Pusey CD. Danger-associated molecular pattern molecules and the receptor for advanced glycation end products enhance ANCA-induced responses. Rheumatology (Oxford) 2021; 61:834-845. [PMID: 33974049 PMCID: PMC8824420 DOI: 10.1093/rheumatology/keab413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/04/2021] [Indexed: 11/25/2022] Open
Abstract
Objectives The pro-inflammatory activities of the calgranulins and HMGB1 can be counteracted by sRAGE, the soluble form of their shared receptor. To understand the role of these molecules in AAV and their potential as therapeutic targets we have studied (i) the relationship between these DAMPS and disease activity; (ii) the expression of RAGE and sRAGE in biopsy tissue and peripheral blood; and (iii) the effect of these molecules on ANCA-mediated cytokine production. Methods We examined circulating levels of calgranulins (S100A8/A9 and S100A12), HMGB1 and sRAGE by ELISA. RAGE was examined in AAV kidney and lung biopsies by immunohistochemistry and RAGE expression was monitored in peripheral blood by qPCR. In vitro, the effect of co-stimulating PBMC with ANCA and S100A8/A9 on cytokine production was studied by ELISA. Results We found significantly raised levels of calgranulins and HMGB1 in active AAV regardless of clinical phenotype (PR3+/MPO+ AAV). Levels of calgranulins showed significant correlations with each other. RAGE protein and message was raised in peripheral blood and in cells infiltrating kidney and lung biopsy tissue, while sRAGE was lowered. Furthermore, ANCA-mediated production of IL-8 from PBMC was significantly enhanced by the presence of S100A8/A9 in a RAGE/TLR4-dependent manner. Conclusions Raised circulating calgranulins provide a good marker of disease activity in AAV and are unlikely to be counteracted by sRAGE. Increased RAGE expression in AAV indicates receptor stimulation in active disease that may exacerbate ANCA-induced cytokine production. Targeting the RAGE pathway may provide a useful therapeutic approach in AAV.
Collapse
Affiliation(s)
- Theresa H Page
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Derick Chiappo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Francesca Brunini
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK.,Nephrology and Dialysis Unit, Ospedale di Circolo e Fondazione Macchi, ASST-Settelaghi, Varese, Italy
| | - Josep Garnica
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK.,Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, 08036, Spain
| | - Jack Blackburn
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Fayaz Dudhiya
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Stephen P McAdoo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
25
|
Yoon T, Ahn SS, Pyo JY, Lee LE, Song JJ, Park YB, Lee SW. Correlation between serum cysteine-rich protein 61 and disease activity of antineutrophil cytoplasmic antibody-associated vasculitis. Clin Rheumatol 2021; 40:3703-3710. [PMID: 33755835 DOI: 10.1007/s10067-021-05701-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/22/2021] [Accepted: 03/15/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Cysteine-rich protein 61 (CYR61) stimulates protein kinase B (Akt)-mediated nuclear factor-kappa B (NF-κB) signalling leading to an increase in pro-inflammatory cytokines, which play important roles in the pathogenesis of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Hence, we investigated whether serum CYR61 was correlated with disease activity of AAV in a single-centre prospective cohort. METHODS Seventy-two patients with AAV were randomly selected and included. Serum CYR61, interleukin (IL)-6 and IL-8 levels were quantified with the patients' stored sera, and clinical and laboratory data at the time of blood sampling were collected. Spearman's correlation and linear regression analysis was conducted to analyse the correlation between continuous variables. The optimal cut-off of serum CYR61 for predicting high disease activity was identified using the receiver operator characteristic curve. Birmingham vasculitis activity score (BVAS) was used as a measure to assess disease activity, and high disease activity was defined as BVAS ≥ 12. RESULTS Serum CYR61 significantly correlated with BVAS (r = 0.249), erythrocyte sedimentation rate (r = 0.283), C-reactive protein (r = 0.298) and serum IL-6 (r = 0.319). However, a linear association was not found between CYR61 and BVAS (β = 0.102, P = 0.304). The relative risk (RR) for high disease activity in AAV patients with serum CYR61 ≥ 236.2 pg/mL was higher than those with serum CYR61 < 236.2 pg/mL (RR 3.316, P = 0.018). CONCLUSION Even though serum CYR61 was not directly proportional to the increase of BVAS, it could be predictive of high disease activity in AAV. Key Points • Serum CYR61 was significantly correlated with BVAS along with ESR, CRP and serum IL-6. • The cut-off of serum CYR61 for high disease activity of AAV was obtained as 236.2 pg/mL. • AAV patients with serum CYR61 ≥ 236.2 pg/mL had increased risk of having higher disease activity than those with serum CYR61 < 236.2 pg/mL (RR 3.316, P = 0.018).
Collapse
Affiliation(s)
- Taejun Yoon
- Department of Medical Science, BK21 Plus Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea
| | - Jung Yoon Pyo
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea
| | - Lucy Eunju Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Seoul, Republic of Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Leacy E, Brady G, Little MA. Pathogenesis of ANCA-associated vasculitis: an emerging role for immunometabolism. Rheumatology (Oxford) 2021; 59:iii33-iii41. [PMID: 32348520 DOI: 10.1093/rheumatology/keaa023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/02/2020] [Indexed: 12/16/2022] Open
Abstract
ANCA-associated vasculitis (AAV) is a severe systemic autoimmune disease. A key feature of AAV is the presence of Anti-Neutrophil Cytoplasmic Antibodies (ANCA) directed against myeloperoxidase (MPO) or proteinase-3 (PR3). ANCA are key to the pathogenesis of AAV, where they activate innate immune cells to drive inflammation. Pre-activation or 'priming' of immune cells appears to be important for complete cellular activation in AAV. The burgeoning field of immunometabolism has illuminated the governance of immune cell function by distinct metabolic pathways. There is ample evidence that the priming events synonymous with AAV alter immune cell metabolism. In this review we discuss the pathogenesis of AAV and its intersection with recent insights into immune cell metabolism.
Collapse
Affiliation(s)
- Emma Leacy
- Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Gareth Brady
- Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Mark A Little
- Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Systemic inflammation response index predicts all-cause mortality in patients with antineutrophil cytoplasmic antibody-associated vasculitis. Int Urol Nephrol 2021; 53:1631-1638. [PMID: 33428165 DOI: 10.1007/s11255-020-02777-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES A systemic inflammation response index (SIRI) has been recently introduced as a tool for the assessment of the prognosis of several critical medical conditions. In this study, we investigated whether SIRI at diagnosis could estimate the cross-sectional disease activity and predict poor prognosis during follow-up in patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). METHODS We reviewed the medical records of 224 immunosuppressive drug-naïve AAV patients and obtained clinical and laboratory data both at diagnosis and during follow-up. SIRI was calculated using the following equation: SIRI = peripheral blood neutrophil count × monocyte count/lymphocyte count. RESULTS The median age of AAV patients at diagnosis was 59.0 years and 33% were male. In the univariable linear regression analysis, SIRI value at diagnosis was not significantly correlated with the cross-sectional Birmingham vasculitis activity score (BVAS) (r = 0.125, P = 0.062). When the SIRI cut-off value at diagnosis was set at 2847.9 mm-3 using the receiver operator characteristic curve, the sensitivity was 56.0% and the specificity was 68.3% for all-cause mortality [area 0.618, 95% confidence interval (CI) 0.502, 0.734]. AAV patients with SIRI ≥ 2847.9 mm-3 had a significantly higher risk for all-cause mortality than those with SIRI < 2847.9 mm-3 [relative risk (RR) 2.747, 95% CI 1.181, 6.392]. During follow-up, AAV patients with SIRI ≥ 2847.9 mm-3 exhibited a significantly lower patients' survival rate than those with SIRI < 2847.9 mm-3 (P = 0.003). CONCLUSIONS SIRI at diagnosis could predict all-cause mortality during follow-up but it could not estimate the cross-sectional BVAS in AAV patients.
Collapse
|
28
|
Immunopathogenesis of ANCA-Associated Vasculitis. Int J Mol Sci 2020; 21:ijms21197319. [PMID: 33023023 PMCID: PMC7584042 DOI: 10.3390/ijms21197319] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis is an autoimmune disorder which affects small- and, to a lesser degree, medium-sized vessels. ANCA-associated vasculitis encompasses three disease phenotypes: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). This classification is largely based on clinical presentations and has several limitations. Recent research provided evidence that genetic background, risk of relapse, prognosis, and co-morbidities are more closely related to the ANCA serotype, proteinase 3 (PR3)-ANCA and myeloperoxidase (MPO)-ANCA, compared to the disease phenotypes GPA or MPA. This finding has been extended to the investigation of biomarkers predicting disease activity, which again more closely relate to the ANCA serotype. Discoveries related to the immunopathogenesis translated into clinical practice as targeted therapies are on the rise. This review will summarize the current understanding of the immunopathogenesis of ANCA-associated vasculitis and the interplay between ANCA serotype and proposed disease biomarkers and illustrate how the extending knowledge of the immunopathogenesis will likely translate into development of a personalized medicine approach in the management of ANCA-associated vasculitis.
Collapse
|
29
|
O'Brien EC, White CA, Wyse J, Leacy E, Porter RK, Little MA, Hickey FB. Pro-inflammatory Stimulation of Monocytes by ANCA Is Linked to Changes in Cellular Metabolism. Front Med (Lausanne) 2020; 7:553. [PMID: 33015103 PMCID: PMC7509421 DOI: 10.3389/fmed.2020.00553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/03/2020] [Indexed: 01/09/2023] Open
Abstract
Clinical and experimental data suggest that pathogenesis in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis is driven by ANCA-mediated activation of neutrophils and monocytes. While the role of neutrophils has been extensively investigated, the function of monocytes remains relatively understudied. We have previously demonstrated that stimulation of monocytes with anti-myeloperoxidase (MPO), but not anti-proteinase-3 (PR3), antibodies results in production of the pro-inflammatory cytokine IL-1β. Changes in cellular metabolism, particularly a switch to glycolysis, have recently been linked to activation of immune cells and production of IL-1β. Therefore, we investigated the metabolic profile of monocytes following ANCA stimulation. We found a significant increase in glucose uptake in anti-MPO stimulated monocytes. Interestingly, both anti-MPO and anti-PR3 stimulation resulted in an immediate increase in glycolysis, measured by Seahorse extracellular flux analysis. However, this increase in glycolysis was sustained (for up to 4 h) in anti-MPO- but not anti-PR3-treated cells. In addition, only anti-MPO-treated cells exhibited increased oxidative phosphorylation, a metabolic response that correlated with IL-1β production. These data indicate that monocyte metabolism is altered by ANCA, with divergent responses to anti-MPO and anti-PR3 antibodies. These metabolic changes may underlie pathologic immune activation in ANCA associated vasculitis, as well as potentially contributing to the differing clinical phenotype between PR3- and MPO-ANCA positive patients. These metabolic pathways may therefore be potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Eóin C O'Brien
- Department of Clinical Medicine, Trinity Health Kidney Centre, Trinity College Dublin, Dublin, Ireland
| | - Carla A White
- Department of Clinical Medicine, Trinity Health Kidney Centre, Trinity College Dublin, Dublin, Ireland
| | - Jason Wyse
- Discipline of Statistics and Information Systems, School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - Emma Leacy
- Department of Clinical Medicine, Trinity Health Kidney Centre, Trinity College Dublin, Dublin, Ireland
| | - Richard K Porter
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Mark A Little
- Department of Clinical Medicine, Trinity Health Kidney Centre, Trinity College Dublin, Dublin, Ireland
| | - Fionnuala B Hickey
- Department of Clinical Medicine, Trinity Health Kidney Centre, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Kitching AR, Anders HJ, Basu N, Brouwer E, Gordon J, Jayne DR, Kullman J, Lyons PA, Merkel PA, Savage COS, Specks U, Kain R. ANCA-associated vasculitis. Nat Rev Dis Primers 2020; 6:71. [PMID: 32855422 DOI: 10.1038/s41572-020-0204-y] [Citation(s) in RCA: 531] [Impact Index Per Article: 106.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
The anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) are a group of disorders involving severe, systemic, small-vessel vasculitis and are characterized by the development of autoantibodies to the neutrophil proteins leukocyte proteinase 3 (PR3-ANCA) or myeloperoxidase (MPO-ANCA). The three AAV subgroups, namely granulomatosis with polyangiitis (GPA), microscopic polyangiitis and eosinophilic GPA (EGPA), are defined according to clinical features. However, genetic and other clinical findings suggest that these clinical syndromes may be better classified as PR3-positive AAV (PR3-AAV), MPO-positive AAV (MPO-AAV) and, for EGPA, by the presence or absence of ANCA (ANCA+ or ANCA-, respectively). Although any tissue can be involved in AAV, the upper and lower respiratory tract and kidneys are most commonly and severely affected. AAVs have a complex and unique pathogenesis, with evidence for a loss of tolerance to neutrophil proteins, which leads to ANCA-mediated neutrophil activation, recruitment and injury, with effector T cells also involved. Without therapy, prognosis is poor but treatments, typically immunosuppressants, have improved survival, albeit with considerable morbidity from glucocorticoids and other immunosuppressive medications. Current challenges include improving the measures of disease activity and risk of relapse, uncertainty about optimal therapy duration and a need for targeted therapies with fewer adverse effects. Meeting these challenges requires a more detailed knowledge of the fundamental biology of AAV as well as cooperative international research and clinical trials with meaningful input from patients.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia. .,Departments of Nephrology and Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia.
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians University, Munich, Germany
| | - Neil Basu
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Elisabeth Brouwer
- Vasculitis Expertise Centre Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Jennifer Gordon
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, PA, USA
| | - David R Jayne
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Paul A Lyons
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Peter A Merkel
- Division of Rheumatology, Department of Medicine and Division of Clinical Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline O S Savage
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Renate Kain
- Department of Pathology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
31
|
Matsumoto K, Suzuki K, Yoshimoto K, Seki N, Tsujimoto H, Chiba K, Takeuchi T. Longitudinal immune cell monitoring identified CD14 ++ CD16 + intermediate monocyte as a marker of relapse in patients with ANCA-associated vasculitis. Arthritis Res Ther 2020; 22:145. [PMID: 32546274 PMCID: PMC7298936 DOI: 10.1186/s13075-020-02234-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Background Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is an autoimmune disease that affects small- to medium-sized blood vessels. Despite treatments having been improved, patients often experience disease relapses. It remains unclear how the immune cells involve in the development of vasculitis and how they fluctuate over the course of treatment. In this study, we aimed to identify the immune subsets and serum cytokines associated with disease relapse by comprehensive immuno-phenotyping in AAV patients. Methods We reviewed consecutive patients (n = 29) from Keio University Hospital who had been newly diagnosed with AAV from January 2015 to February 2019 and chronologically followed until 52 weeks. Numbers of circulating T cells, B cells, monocytes, and granulocytes were analyzed by flow cytometry (FACS). Serum levels of cytokines were measured by electrochemiluminescence enzyme immunoassay. Clinical information was obtained from patients’ records and association with time-course changes in immuno-phenotypes and serum levels of cytokines were assessed. Results Comprehensive immuno-phenotyping data from 161 samples from 29 AAV patients at diagnosis; at weeks 4, 12, 24, and 52 of treatment; and at time of major relapse were examined. FACS analysis from patients with relapse revealed that CD14++ CD16+ intermediate monocytes and plasma cells concomitantly changed associated with disease relapse, which were independent from treatment regimen, ANCA status, or disease phenotype. In particular, the number of CD14++ CD16+ intermediate monocytes at relapse was significantly higher than that in remission or in healthy controls. Serum cytokine measurement revealed that changes of monocyte-derived proinflammatory cytokines such as IL-1β, IL-6, IL-8, and TNF-α were associated with disease status. Conclusions Chronological changes in CD14++ CD16+ intermediate monocyte counts can be a marker of disease relapse in AAV patients.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Keiko Yoshimoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.,Clinical and Translational Research Center, Keio University Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Noriyasu Seki
- Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Hideto Tsujimoto
- Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Kenji Chiba
- Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
32
|
Torres-Ruiz J, Carrillo-Vazquez DA, Padilla-Ortiz DM, Vazquez-Rodriguez R, Nuñez-Alvarez C, Juarez-Vega G, Gomez-Martin D. TLR expression in peripheral monocyte subsets of patients with idiopathic inflammatory myopathies: association with clinical and immunological features. J Transl Med 2020; 18:125. [PMID: 32164729 PMCID: PMC7066841 DOI: 10.1186/s12967-020-02290-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Monocytes and toll-like receptors (TLR) have been found in the inflammatory infiltrate of muscle biopsies in patients with idiopathic inflammatory myopathies (IIM), suggesting an important role of these cells in the pathogenesis of myositis. The monocyte subsets, their TLR expression in peripheral blood and their relationship with the clinical characteristics of patients with IIM has not been addressed. METHODS We recruited 45 patients with IIM diagnosis and 15 age and sex-adjusted healthy controls. We assessed the disease activity and damage, performed a nailfold capillaroscopy and registered the cardio-pulmonary parameters from the medical charts. Monocyte subsets, their expression of TLR2 and TLR4 and the serum Th1/Th2/Th17 cytokines levels were evaluated by flow cytometry. We expressed quantitative variables as medians and interquartile ranges (IQR) or minimum and maximum (min-max). Differences between groups were assessed with Mann-Whitney U and the Kruskal-Wallis tests. Correlation between quantitative variables was assessed with Spearman Rho. RESULTS Twenty-nine patients were women (64.4%) and 32 (71.1%) had dermatomyositis. In comparison to healthy controls, patients with active IIM had a higher percentage of intermediate monocytes and lower amounts of classical monocytes. Patients with IIM had a higher expression of TLR4 in all their monocyte subsets, regardless of disease activity and prednisone treatment. Serum IL-6 correlated with the TLR2 expression in every monocyte subset and the expression of TLR2 in intermediate monocytes was higher among patients with dysphagia. Subjects with nailfold capillaroscopy abnormalities had a higher amount of TLR2+ classical and non-classical monocytes and those with interstitial lung disease (ILD) had a higher percentage of TLR4+ non-classical monocytes. The classical and intermediate monocytes from patients with anti Mi2 antibodies had a higher expression of TLR4. The percentage of intermediate monocytes and the expression of TLR4 in all monocyte subsets showed a good diagnostic capacity in patients with IIM. CONCLUSION Patients with IIM have a differential pool of monocyte subsets with an enhanced expression of TLR2 and TLR4, which correlates with disease activity and distinctive clinical features including dysphagia, ILD, vasculopathy, and pro-inflammatory cytokines. These immunological features might be useful as a potential diagnostic tool as well as novel disease activity biomarkers in IIM.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Emergency Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Alberto Carrillo-Vazquez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Marcela Padilla-Ortiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Universidad de La Sabana, Hospital Militar Central, Bogotá, DC, Colombia
| | - Ricardo Vazquez-Rodriguez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Nuñez-Alvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guillermo Juarez-Vega
- Flow Cytometry Unit, Red de Apoyo a la Investigación, Coordinación de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Gomez-Martin
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave Morones Prieto 3000, 64710, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
33
|
Salama AD. Genetics and pathogenesis of small-vessel vasculitis. Best Pract Res Clin Rheumatol 2018; 32:21-30. [PMID: 30526895 DOI: 10.1016/j.berh.2018.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Small-vessel vasculitides are uncommon autoimmune diseases characterised by inflammation and necrosis of arterioles, capillaries and venules, frequently described as various (previously eponymous) clinical syndromes. Some are associated with vessel wall immune complex deposition, whereas others are pauci-immune but paradoxically often associated with circulating anti-neutrophil cytoplasmic antibodies (ANCA). Most is known about the pathogenesis of the pauci-immune ANCA-associated syndromes, which are gradually becoming better understood with regard to their genetic predisposition and the critical pathways mediating disease initiation, as well as their particular phenotypic features. Through better understanding of key cellular and molecular players, we have been able to develop novel biomarkers and treatment strategies, which should translate to improved diagnostics, treatment protocols and, ultimately, better patient outcomes. These conditions are treatable but not yet curable, although it is clear that patients may follow different disease courses, which for some include restoration of their pre-morbid immune status.
Collapse
Affiliation(s)
- Alan D Salama
- UCL Department of Renal Medicine, Centre for Experimental Nephrology, Royal Free Hospital, London, NW3 2PF, United Kingdom.
| |
Collapse
|
34
|
van der Geest KSM, Brouwer E, Sanders JS, Sandovici M, Bos NA, Boots AMH, Abdulahad WH, Stegeman CA, Kallenberg CGM, Heeringa P, Rutgers A. Towards precision medicine in ANCA-associated vasculitis. Rheumatology (Oxford) 2018; 57:1332-1339. [PMID: 29045715 DOI: 10.1093/rheumatology/kex367] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Indexed: 12/18/2022] Open
Abstract
ANCA-associated vasculitis (AAV) is characterized by inflammation and destruction of small and medium-sized vessels. Current management strategies for AAV have been validated in large groups of patients. However, recent insights indicate that distinct patient subsets may actually exist within AAV, thereby justifying the development of more personalized treatment strategies. In this review, we discuss current evidence for a better classification of AAV based on ANCA type. We describe how thus defined categories of AAV patients may differ in genetic background, clinical presentation, immune pathology, response to treatment and disease outcome. We also explore how these insights may provide a rationale for targeted treatments in different categories of AAV patients. Finally, we provide recommendations on how to further establish precision medicine in AAV.
Collapse
Affiliation(s)
- Kornelis S M van der Geest
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Brouwer
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Stephan Sanders
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria Sandovici
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nicolaas A Bos
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annemieke M H Boots
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wayel H Abdulahad
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Coen A Stegeman
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cees G M Kallenberg
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Heeringa
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Abraham Rutgers
- Vasculitis Expertise Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
35
|
Hutton HL, Holdsworth SR, Kitching AR. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin Nephrol 2018; 37:418-435. [PMID: 28863790 DOI: 10.1016/j.semnephrol.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Our understanding of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis has developed greatly since the discovery of ANCA, directed against neutrophil components, in 1982. Observations in human disease, and increasingly sophisticated studies in vitro and in rodent models in vivo, have allowed a nuanced understanding of many aspects of the immunopathogenesis of disease, including the significance of ANCA as a diagnostic and monitoring tool as well as a mediator of microvascular injury. The mechanisms of leukocyte recruitment and tissue injury, and the role of T cells increasingly are understood. Unexpected findings, such as the role of complement, also have been uncovered through experimental studies and human observations. This review focusses on the pathogenesis of ANCA-associated vasculitis, highlighting the challenges in finding new, less-toxic treatments and potential therapeutic targets in this disease. The current suite of rodent models is reviewed, and future directions in the study of this complex and fascinating disease are suggested.
Collapse
Affiliation(s)
- Holly L Hutton
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Pediatric Nephrology, Monash Children's Hospital, Clayton, Victoria, Australia.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The ANCA-associated vasculitides are a group of small vessel vasculitides characterized by autoantibodies recognizing the neutrophil cytoplasmic antigens PR3 and MPO. We examine the current clinical and molecular immunology understanding of ANCA-associated vasculitides and discuss the current needs in our understanding of the pathogenic mechanisms of these rare diseases. RECENT FINDINGS The majority of efforts to understand the pathogenesis of these diseases have focused on dissecting neutrophil biology because the neutrophil is the primary expressor of ANCA autoantigens. However, a number of important genetic, clinical, and cellular biology observations suggest that attempts to understand the pathogenesis of ANCA vasculitides should move away from emphasis on the role of the neutrophil and instead re-focus on the potential role of other immune cell mediators. Whether or not neutrophils are the key determinant of ANCA-associated vasculitis pathogenesis should be revisited in detail. A neutrophil-centric view of the pathogenesis of these diseases cannot fully account for important genetic, clinical, and cellular biology observations that implicate important and under-appreciated roles for monocytes and T cells. Refocusing on these findings will likely lead to new discovery of novel therapeutic targets and the identification of clinically useful biomarkers for disease activity.
Collapse
Affiliation(s)
- Eric J Gapud
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, MFL Center Tower, Ste. 5300, Baltimore, MD, 21224, USA
| | - Philip Seo
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, MFL Center Tower, Ste. 5300, Baltimore, MD, 21224, USA
| | - Brendan Antiochos
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, MFL Center Tower, Ste. 5300, Baltimore, MD, 21224, USA.
| |
Collapse
|
37
|
Monocytes Promote Crescent Formation in Anti-Myeloperoxidase Antibody–Induced Glomerulonephritis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1908-1915. [DOI: 10.1016/j.ajpath.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 11/20/2022]
|
38
|
Involvement of Monocyte Subsets in the Immunopathology of Giant Cell Arteritis. Sci Rep 2017; 7:6553. [PMID: 28747747 PMCID: PMC5529580 DOI: 10.1038/s41598-017-06826-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022] Open
Abstract
Monocytes/macrophages are critical in systemic and local inflammation in giant cell arteritis (GCA) and possibly in clinically overlapping polymyalgia rheumatica (PMR). Therefore, we aimed to understand the contribution of monocyte subsets and the CX3CR1-CX3CL1 and CCR2-CCL2 migratory pathways, to the pathology of GCA. Peripheral blood monocytes were enumerated in samples from newly-diagnosed, untreated GCA and PMR patients and after prednisone-induced remission. The distribution of classical (CD14brightCD16neg) and the more pro-inflammatory, intermediate (CD14brightCD16+) and non-classical (CD14dimCD16+) monocyte subsets was analysed by flow cytometry. The phenotype of macrophages in temporal artery biopsies (TABs) from GCA patients was studied by immunohistochemistry and immunofluorescence. A clear monocytosis was seen in newly diagnosed GCA and PMR patients caused by elevated numbers of classical monocytes. Prednisone treatment suppressed numbers of non-classical monocytes. Both chemokine CX3CL1 and CCL2 were highly expressed in the TAB. Most macrophages in the TAB of GCA patients expressed non-classical monocyte markers CD16 and CX3CR1 whereas co-localisation of CD16 with classical monocyte marker CCR2 was infrequent. In conclusion, we report an altered distribution of monocyte subsets in both GCA and PMR patients. The majority of macrophages in TABs of GCA patients were CD68 + CD16 + CX3CR1 + CCR2- and thereby resembled the phenotype of non-classical monocytes.
Collapse
|
39
|
Caster DJ, Powell DW, Miralda I, Ward RA, McLeish KR. Re-Examining Neutrophil Participation in GN. J Am Soc Nephrol 2017; 28:2275-2289. [PMID: 28620081 DOI: 10.1681/asn.2016121271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Significant advances in understanding the pathogenesis of GN have occurred in recent decades. Among those advances is the finding that both innate and adaptive immune cells contribute to the development of GN. Neutrophils were recognized as key contributors in early animal models of GN, at a time when the prevailing view considered neutrophils to function as nonspecific effector cells that die quickly after performing antimicrobial functions. However, advances over the past two decades have shown that neutrophil functions are more complex and sophisticated. Specifically, research has revealed that neutrophil survival is regulated by the inflammatory milieu and that neutrophils demonstrate plasticity, mediate microbial killing through previously unrecognized mechanisms, demonstrate transcriptional activity leading to the release of cytokines and chemokines, interact with and regulate cells of the innate and adaptive immune systems, and contribute to the resolution of inflammation. Therefore, neutrophil participation in glomerular diseases deserves re-evaluation. In this review, we describe advances in understanding classic neutrophil functions, review the expanded roles of neutrophils in innate and adaptive immune responses, and summarize current knowledge of neutrophil contributions to GN.
Collapse
Affiliation(s)
- Dawn J Caster
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, .,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| | - David W Powell
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Irina Miralda
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Richard A Ward
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kenneth R McLeish
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky.,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| |
Collapse
|
40
|
Popat RJ, Hakki S, Thakker A, Coughlan AM, Watson J, Little MA, Spickett CM, Lavender P, Afzali B, Kemper C, Robson MG. Anti-myeloperoxidase antibodies attenuate the monocyte response to LPS and shape macrophage development. JCI Insight 2017; 2:e87379. [PMID: 28138552 PMCID: PMC5256146 DOI: 10.1172/jci.insight.87379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) vasculitis is characterized by the presence of autoantibodies to myeloperoxidase and proteinase-3, which bind monocytes in addition to neutrophils. While a pathological effect on neutrophils is acknowledged, the impact of ANCA on monocyte function is less well understood. Using IgG from patients we investigated the effect of these autoantibodies on monocytes and found that anti-myeloperoxidase antibodies (MPO-ANCA) reduced both IL-10 and IL-6 secretion in response to LPS. This reduction in IL-10 and IL-6 depended on Fc receptors and enzymatic myeloperoxidase and was accompanied by a significant reduction in TLR-driven signaling pathways. Aligning with changes in TLR signals, oxidized phospholipids, which function as TLR4 antagonists, were increased in monocytes in the presence of MPO-ANCA. We further observed that MPO-ANCA increased monocyte survival and differentiation to macrophages by stimulating CSF-1 production. However, this was independent of myeloperoxidase enzymatic activity and TLR signaling. Macrophages differentiated in the presence of MPO-ANCA secreted more TGF-β and further promoted the development of IL-10– and TGF-β–secreting CD4+ T cells. Thus, MPO-ANCA may promote inflammation by reducing the secretion of antiinflammatory IL-10 from monocytes, and MPO-ANCA can alter the development of macrophages and T cells to potentially promote fibrosis. Anti-myeloperoxidase antibodies from patients with anti-neutrophil cytoplasmic antibody (ANCA) vasculitis alter monocyte function in addition to previously described effects on neutrophils.
Collapse
Affiliation(s)
- Reena J Popat
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Seran Hakki
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Alpesh Thakker
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom
| | - Alice M Coughlan
- Trinity Health Kidney Centre, Department of Clinical Medicine, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland
| | - Julie Watson
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Mark A Little
- Trinity Health Kidney Centre, Department of Clinical Medicine, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland
| | - Corinne M Spickett
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom
| | - Paul Lavender
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Behdad Afzali
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| | - Michael G Robson
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London, United Kingdom
| |
Collapse
|
41
|
Bessler WK, Hudson FZ, Zhang H, Harris V, Wang Y, Mund JA, Downing B, Ingram DA, Case J, Fulton DJ, Stansfield BK. Neurofibromin is a novel regulator of Ras-induced reactive oxygen species production in mice and humans. Free Radic Biol Med 2016; 97:212-222. [PMID: 27266634 PMCID: PMC5765860 DOI: 10.1016/j.freeradbiomed.2016.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/25/2016] [Accepted: 06/02/2016] [Indexed: 12/22/2022]
Abstract
Neurofibromatosis type 1 (NF1) predisposes individuals to early and debilitating cardiovascular disease. Loss of function mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin, leads to accelerated p21(Ras) activity and phosphorylation of multiple downstream kinases, including Erk and Akt. Nf1 heterozygous (Nf1(+/-)) mice develop a robust neointima that mimics human disease. Monocytes/macrophages play a central role in NF1 arterial stenosis as Nf1 mutations in myeloid cells alone are sufficient to reproduce the enhanced neointima observed in Nf1(+/-) mice. Though the molecular mechanisms underlying NF1 arterial stenosis remain elusive, macrophages are important producers of reactive oxygen species (ROS) and Ras activity directly regulates ROS production. Here, we use compound mutant and lineage-restricted mice to demonstrate that Nf1(+/-) macrophages produce excessive ROS, which enhance Nf1(+/-) smooth muscle cell proliferation in vitro and in vivo. Further, use of a specific NADPH oxidase-2 inhibitor to limit ROS production prevents neointima formation in Nf1(+/-) mice. Finally, mononuclear cells from asymptomatic NF1 patients have increased oxidative DNA damage, an indicator of chronic exposure to oxidative stress. These data provide genetic and pharmacologic evidence that excessive exposure to oxidant species underlie NF1 arterial stenosis and provide a platform for designing novels therapies and interventions.
Collapse
Affiliation(s)
- Waylan K Bessler
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Pediatrics and Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis 46202, United States
| | - Farlyn Z Hudson
- Department of Pediatrics and Neonatal-Perinatal Medicine, Augusta University, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States
| | - Hanfang Zhang
- Department of Pediatrics and Neonatal-Perinatal Medicine, Augusta University, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States
| | - Valerie Harris
- Department of Pediatrics and Neonatal-Perinatal Medicine, Augusta University, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States
| | - Yusi Wang
- Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, United States
| | - Julie A Mund
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Pediatrics and Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis 46202, United States; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis 46202, United States
| | - Brandon Downing
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Pediatrics and Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis 46202, United States
| | - David A Ingram
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Pediatrics and Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis 46202, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis 46202, United States
| | - Jamie Case
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis 46202, United States; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis 46202, United States; Scripps Clinic Medical Group, Center for Organ and Cell Transplantation, La Jolla, CA 92037, United States
| | - David J Fulton
- Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, United States
| | - Brian K Stansfield
- Department of Pediatrics and Neonatal-Perinatal Medicine, Augusta University, Augusta, GA 30912, United States; Vascular Biology Center, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
42
|
Jarrot PA, Kaplanski G. Pathogenesis of ANCA-associated vasculitis: An update. Autoimmun Rev 2016; 15:704-13. [DOI: 10.1016/j.autrev.2016.03.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 01/17/2023]
|