1
|
Tian Q, Li Z, Yan Z, Jiang S, Zhao X, Wang L, Li M. Inflammatory role of S100A8/A9 in the central nervous system non-neoplastic diseases. Brain Res Bull 2024; 218:111100. [PMID: 39396712 DOI: 10.1016/j.brainresbull.2024.111100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
S100A8 (MRP8) and S100A9 (MRP14) are critical mediators of the inflammatory response; they are usually present as heterodimers because of the instability of homodimers. Studies have demonstrated that S100A8/A9 expression is significantly upregulated in several central nervous system (CNS) diseases. S100A8/A9 is actively released by neutrophils and monocytes; it plays a key role in regulating the inflammatory response by stimulating leukocyte recruitment and inducing cytokine secretion during inflammation. Additionally, S100A8/A9 can be used as a diagnostic biomarker for several CNS diseases and as a predictor of therapeutic response to inflammation-related diseases. In this work, we reviewed our current understanding of S100A8/A9 overexpression in inflammation and its importance in the development and progression of CNS inflammatory diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and stroke, and the functional roles and therapeutic applications of S100A8/A9 in these diseases. Finally, we discussed the current barriers and future research directions of S100A8/A9 in CNS diseases.
Collapse
Affiliation(s)
- Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Zhijie Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Ziang Yan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Shengming Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Xincan Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Lei Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei 443000, China; Department of Neurosurgery, Yichang Central People's Hospital, Yichang, Hubei, China.
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
2
|
Rosner GM, Goswami HB, Sessions K, Mendyka LK, Kerin B, Vlasac I, Mellinger D, Gwilt L, Hampton TH, Graber M, Ashare A, Harris WT, Christensen B, Stanton BA, Swiatecka-Urban A, Skopelja-Gardner S. Lung-Kidney Axis in Cystic Fibrosis: Early Urinary Markers of Kidney Injury Correlate with Neutrophil Activation and Worse Lung Function. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.11.10.23298378. [PMID: 39371147 PMCID: PMC11451629 DOI: 10.1101/2023.11.10.23298378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Adult people with cystic fibrosis (PwCF) have a higher risk of end-stage kidney disease than the general population. The nature and mechanism of kidney disease in CF are unknown. This study quantifies urinary kidney injury markers and examines the hypothesis that neutrophil activation and lung infection are associated with early kidney injury in CF. Methods Urinary total protein, albumin, and markers of kidney injury and neutrophil activation, normalized to creatinine, as well as urinary immune cells, were quantified in CF (n = 48) and healthy (n = 33) cohorts. Infection burden and chronicity were defined by sputum culture and serum titers of anti-bacterial antibodies. Results PwCF had increased urinary protein levels, consisting of low-molecular-weight tubular injury markers, independent of glomerular filtration rate (eGFR). This finding suggests subclinical renal injury processes. Urinary analysis of the CF cohort identified different associations of urinary injury markers with aminoglycoside exposure, lung function, and neutrophil activation. High urinary KIM-1 levels and increased prevalence of neutrophils among urine immune cells correlated with decreased lung function in PwCF. The relationship between tubular injury and decreased lung function was most prominent in patients harboring chronic Pseudomonas aeruginosa infection. Conclusions Increased urinary tubular injury markers in PwCF suggest early subclinical renal injury not readily detected by eGFR. The strong association of high urinary KIM-1 and neutrophils with diminished lung function and high Pseudomonas aeruginosa burden suggests that pulmonary disease may contribute to renal injury in CF.
Collapse
Affiliation(s)
- Grace M. Rosner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Himanshu B. Goswami
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Katherine Sessions
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Lindsay K. Mendyka
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Brenna Kerin
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Irma Vlasac
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Diane Mellinger
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Lorraine Gwilt
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Thomas H. Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Martha Graber
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Alix Ashare
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Department of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | | | - Brock Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Bruce A. Stanton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Sladjana Skopelja-Gardner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
3
|
Chen S, Fan Y, Wu Q, Zhang G, Wang Y, Li W, Yang S, Matucci-Cerinic M, Furst DE. Integrative Transcriptomic Analysis of Peripheral Blood Monocytes in Systemic Sclerosis and Shared Pathogenic Pathways in Autoimmune Diseases. Arch Med Res 2024; 56:103072. [PMID: 39208548 DOI: 10.1016/j.arcmed.2024.103072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/04/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Systemic sclerosis (SSc) is an autoimmune disease (AD), that receives less attention compared to rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and primary Sjögren's syndrome (pSS). This study aims to analyze transcriptional profiles and immune cell composition in peripheral blood mononuclear cells (PBMC) from SSc patients compared to other ADs. METHODS RNA-seq data from 119 untreated patients (eight with SSc, 42 with RA, 41 with pSS, 28 with SLE) and 20 healthy controls were analyzed. Bioinformatics tools were employed to identify differentially expressed genes (DEGs), biological functions and immune cell profiles unique to SSc and shared with other ADs. RESULTS 1,148 DEGs were found in SSc, with upregulated genes associated with megakaryocyte processes and downregulated genes associated with neutrophil function and immune response. DEGs, including ALDH1A1 and MEGF9, were associated with neutropenia. Upregulated transcription factors (TFs) were linked to embryonic hematopoiesis and downregulated TFs were involved in leukocyte differentiation and immune regulation. Comparative analysis with other ADs revealed common pathogenic pathways, emphasizing megakaryocyte proliferation. Neutrophils count was significantly decreased in ADs (p < 0.001) compared to healthy controls. Comparative analysis highlighted common pathways, particularly in megakaryocyte proliferation, and unique genes (MEGF9, MMP8, and KRT family members) in SSc, suggesting roles in neutrophil function, skin integrity, and fibrosis. CONCLUSIONS This study identifies dysregulated gene expression (KRT and MMP8) associated with neutrophil function and increased megakaryocytes in SSc, highlighting common patterns across autoimmune diseases. These findings offer new insights into the potential pathogenesis of SSc, and help to explore new targets for the treatment.
Collapse
Affiliation(s)
- Shaoqi Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yu Fan
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Qiulin Wu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yukai Wang
- Department of Rheumatology and Immunology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Weiping Li
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Shengli Yang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China; Shanghai Academician Consulting and Academic Activities Center of Chinese Academy of Engineering, Shanghai, China.
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, San Raffaele Hospital, Milan, Italy
| | - Daniel E Furst
- Division of Rheumatology, School of Medicine, University of California at Los Angeles, California, USA
| |
Collapse
|
4
|
Chen J, Cao Y, Xiao J, Hong Y, Zhu Y. The emerging role of neutrophil extracellular traps in the progression of rheumatoid arthritis. Front Immunol 2024; 15:1438272. [PMID: 39221253 PMCID: PMC11361965 DOI: 10.3389/fimmu.2024.1438272] [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: 05/25/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. Neutrophil extracellular traps (NETs are NETwork protein structures activated by neutrophils to induce the cleavage and release of DNA-protein complexes). Current studies have shown the critical involvement of NETs in the progression of autoimmune diseases, Neutrophils mostly gather in the inflammatory sites of patients and participate in the pathogenesis of autoimmune diseases in various ways. NETs, as the activated state of neutrophils, have attracted much attention in immune diseases. Many molecules released in NETs are targeted autoantigens in autoimmune diseases, such as histones, citrulline peptides, and myeloperoxidase. All of these suggest that NETs have a direct causal relationship between the production of autoantigens and autoimmune diseases. For RA in particular, as a disorder of the innate and adaptive immune response, the pathogenesis of RA is inseparable from the generation of RA. In this article, we investigate the emerging role of NETs in the pathogenesis of RA and suggest that NETs may be an important target for the treatment of inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Jingjing Chen
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yang Cao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Jing Xiao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yujie Hong
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yan Zhu
- The Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
5
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024. [PMID: 39105461 DOI: 10.1002/ptr.8311] [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: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Wang T, Rathee A, Pemberton PA, Lood C. Exogenous serpin B1 restricts immune complex-mediated NET formation via inhibition of a chymotrypsin-like protease and enhances microbial phagocytosis. J Biol Chem 2024; 300:107533. [PMID: 38971315 PMCID: PMC11327461 DOI: 10.1016/j.jbc.2024.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024] Open
Abstract
Immune complex (IC)-driven formation of neutrophil extracellular traps (NETs) is a major contributing factor to the pathogenesis of autoimmune diseases including systemic lupus erythematosus (SLE). Exogenous recombinant human serpin B1 (rhsB1) can regulate NET formation; however, its mechanism(s) of action is currently unknown as is its ability to regulate IC-mediated NET formation and other neutrophil effector functions. To investigate this, we engineered or post-translationally modified rhsB1 proteins that possessed specific neutrophil protease inhibitory activities and pretreated isolated neutrophils with them prior to inducing NET formation with ICs derived from patients with SLE, PMA, or the calcium ionophore A23187. Neutrophil activation and phagocytosis assays were also performed with rhsB1 pretreated and IC-activated neutrophils. rhsB1 dose-dependently inhibited NET formation by all three agents in a process dependent on its chymotrypsin-like inhibitory activity, most likely cathepsin G. Only one variant (rhsB1 C344A) increased surface levels of neutrophil adhesion/activation markers on IC-activated neutrophils and boosted intracellular ROS production. Further, rhsB1 enhanced complement-mediated neutrophil phagocytosis of opsonized bacteria but not ICs. In conclusion, we have identified a novel mechanism of action by which exogenously administered rhsB1 inhibits IC, PMA, and A2138-mediated NET formation. Cathepsin G is a well-known contributor to autoimmune disease but to our knowledge, this is the first report implicating it as a potential driver of NET formation. We identified the rhsB1 C334A variant as a candidate protein that can suppress IC-mediated NET formation, boost microbial phagocytosis, and potentially impact additional neutrophil effector functions including ROS-mediated microbial killing in phagolysosomes.
Collapse
Affiliation(s)
- Ting Wang
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Arpit Rathee
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Christian Lood
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
7
|
Frade-Sosa B, Ponce A, Ruiz-Ortiz E, De Moner N, Gómara MJ, Azuaga AB, Sarmiento-Monroy JC, Morlà R, Ruiz-Esquide V, Macías L, Sapena N, Tobalina L, Ramirez J, Cañete JD, Yague J, Auge JM, Gomez-Puerta JA, Viñas O, Haro I, Sanmarti R. Neutrophilic Activity Biomarkers (Plasma Neutrophil Extracellular Traps and Calprotectin) in Established Patients with Rheumatoid Arthritis Receiving Biological or JAK Inhibitors: A Clinical and Ultrasonographic Study. Rheumatol Ther 2024; 11:501-521. [PMID: 38430455 PMCID: PMC11111434 DOI: 10.1007/s40744-024-00650-9] [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: 11/15/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024] Open
Abstract
INTRODUCTION This study assesses the accuracy of neutrophil activation markers, including neutrophil extracellular traps (NETs) and calprotectin, as biomarkers of disease activity in patients with established rheumatoid arthritis (RA). We also analyse the relationship between NETs and various types of therapies as well as their association with autoimmunity. METHODS Observational cross-sectional study of patients with RA receiving treatment with biological disease-modifying antirheumatic drugs or Janus kinase inhibitors (JAK-inhibitors) for at least 3 months. Plasma calprotectin levels were measured using an enzyme-linked immunosorbent assay test kit and NETs by measuring their remnants in plasma (neutrophil elastase-DNA and histone-DNA complexes). We also assessed clinical disease activity, joint ultrasound findings and autoantibody status [reumatoid factor (RF), anti-citrullinated peptide/protein antibodies (ACPAs) and anti-carbamylated protein (anti-CarP)]. Associations between neutrophilic biomarkers and clinical or ultrasound scores were sought using correlation analysis. The discriminatory capacity of both neutrophilic biomarkers to detect ultrasound synovitis was analysed through receiver-operating characteristic (ROC) curves. RESULTS One hundred fourteen patients were included. Two control groups were included to compare NET levels. The active control group consisted of 15 patients. The second control group consisted of 30 healthy subjects. Plasma NET levels did not correlate with clinical disease status, regardless of the clinic index analysed or the biological therapy administered. No significant correlation was observed between NET remnants and ultrasound synovitis. There was no correlation between plasma NET and autoantibodies. In contrast, plasma calprotectin positively correlated with clinical parameters (swollen joint count [SJC] rho = 0.49; P < 0.001, Clinical Disease Activity Index [CDAI] rho = 0.30; P < 0.001) and ultrasound parameters (rho > 0.50; P < 0.001). Notably, this correlation was stronger than that observed with acute phase reactants. CONCLUSION While NET formation induced by neutrophils may play a role in RA pathogenesis, our study raises questions about the utility of NET remnants in peripheral circulation as a biomarker for inflammatory activity. In contrast, this study strongly supports the usefulness of calprotectin as a biomarker of inflammatory activity in patients with RA.
Collapse
Affiliation(s)
- Beatriz Frade-Sosa
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Andrés Ponce
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Estíbaliz Ruiz-Ortiz
- Department of Immunology-CDB, Hospital Clinic of Barcelona, Barcelona, Barcelona, Spain
| | - Noemí De Moner
- Department of Immunology-CDB, Hospital Clinic of Barcelona, Barcelona, Barcelona, Spain
| | - María J Gómara
- Unit of Synthesis and Biomedical Applications of Peptides, Institute for Advanced Chemistry of Catalonia. Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Ana Belén Azuaga
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Juan C Sarmiento-Monroy
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Rosa Morlà
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Virginia Ruiz-Esquide
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Laura Macías
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Nuria Sapena
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Lola Tobalina
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Julio Ramirez
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Juan D Cañete
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Jordi Yague
- Department of Immunology-CDB, Hospital Clinic of Barcelona, Barcelona, Barcelona, Spain
| | - Josep M Auge
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - José A Gomez-Puerta
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain
| | - Odette Viñas
- Department of Immunology-CDB, Hospital Clinic of Barcelona, Barcelona, Barcelona, Spain
| | - Isabel Haro
- Unit of Synthesis and Biomedical Applications of Peptides, Institute for Advanced Chemistry of Catalonia. Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Raimon Sanmarti
- Department of Rheumatology, Hospital Clinic of Barcelona, Barcelona, IDIBAPS, Carrer Villarroel 170, 08170, Barcelona, Spain.
| |
Collapse
|
8
|
Mao J, Tan M, Li J, Liu C, Hao J, Zheng J, Shen H. Neutrophil Extracellular Traps Induce Pyroptosis of Rheumatoid Arthritis Fibroblast-Like Synoviocytes via the NF-κB/Caspase 3/GSDME Pathway. Inflammation 2024; 47:921-938. [PMID: 38133702 DOI: 10.1007/s10753-023-01951-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/23/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Rheumatoid arthritis (RA) is an enduring, progressive autoimmune disorder. Abnormal activation of fibroblast-like synoviocytes (FLSs) has been proposed as the initiating factor for inflammation of the synovium and bone destruction. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA, histones, and granule proteins, are involved in the development of RA in multiple aspects. Pyroptosis, gasdermin-mediated inflammatory programmed cell death, plays a vital function in the etiopathogenesis of RA. However, the exact mechanism underlying NETs-induced pyroptosis in FLSs of RA and its impact on cellular pathogenic behavior remain undefined. In this study, we demonstrated that gasdermin E (GSDME) expression was upregulated in RA plasma and synoviums, which was positively correlated with the elevated cell-free DNA (cfDNA) and citrullinated histone 3 (Cit H3) levels in the plasma. Additionally, in vitro experiments have shown that NETs triggered caspase 3/GSDME-mediated pyroptosis in RA-FLSs, characterized by decreased cell viability, cell membrane blebbing, and rupture, as well as increased levels of pyroptosis-related proteins and pro-inflammatory cytokines. Again, silencing GSDME significantly inhibited pyroptosis and suppressed the migration, invasion, and secretion of pro-inflammatory cytokines in RA-FLSs. Furthermore, we also found that the nuclear factor-kappa B (NF-κB) pathway, serving as an upstream mechanism, was involved in FLS pyroptosis. In conclusion, our investigation indicated that NETs could induce RA-FLS pyroptosis and facilitate phenotypic transformation through targeting the NF-κB/caspase 3/GSDME axis. This is the first to explore the crucial role of NETs-induced FLS pyroptosis in the progression of RA, providing novel targets for the clinical management of refractory RA.
Collapse
Affiliation(s)
- Jing Mao
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Min Tan
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jun Li
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Chunhua Liu
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jiayao Hao
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jianxiong Zheng
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Haili Shen
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
9
|
Carmona-Rivera C, Kaplan MJ, O'Neil LJ. Neutrophils in Inflammatory Bone Diseases. Curr Osteoporos Rep 2024; 22:280-289. [PMID: 38418800 PMCID: PMC11061041 DOI: 10.1007/s11914-024-00865-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE OF REVIEW In this review, we summarize the current evidence that suggests that neutrophils play a key role in facilitating damage to local bone structures. RECENT FINDINGS Neutrophil infiltration is a hallmark of inflammatory bone diseases such as rheumatoid arthritis (RA) and periodontitis disease (PD). Both of these human diseases are marked by an imbalance in bone homeostasis, favoring the degradation of local bone which ultimately leads to erosions. Osteoclasts, a multinucleated resident bone cell, are responsible for facilitating the turnover of bone and the bone damage observed in these diseases. The involvement of neutrophils and neutrophil extracellular trap formation have recently been implicated in exacerbating osteoclast function through direct and indirect mechanisms. We highlight a recent finding that NET proteins such as histones and elastase can generate non-canonical, inflammatory osteoclasts, and this process is mediated by post-translational modifications such as citrullination and carbamylation, both of which act as autoantigens in RA. It appears that NETs, autoantibodies, modified proteins, cytokines, and osteoclasts all ultimately contribute to local and permanent bone damage in RA and PD. However, more studies are needed to fully understand the role of neutrophils in inflammatory bone diseases.
Collapse
Affiliation(s)
- Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Liam J O'Neil
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
10
|
Xv Z, Xv X, Chen N, Yuan J, Li J, Wang L, Yu S, Li G, Ding M, Zong M, Fan L. Soluble signal inhibitory receptor on leukocytes-1 reflects disease activity and assists diagnosis of patients with rheumatoid arthritis. Clin Chim Acta 2024; 556:117808. [PMID: 38309555 DOI: 10.1016/j.cca.2024.117808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND SIRL-1, an immunosuppressive receptor encoded by the VSTM1 gene, has recently been linked to rheumatoid arthritis (RA) due to its association with activated polymorphonuclear neutrophils (PMNs). Considering that the activated PMNs play a crucial role in the pathogenesis of rheumatoid arthritis (RA), we aimed to measure the levels of soluble SIRL-1, investigating whether they add value to RA in the clinical diagnosis. METHODS Utilizing an enzyme-linked immunosorbent assay, the concentration of sSIRL-1 was measured in serum samples from cohort 1 diagnosed with RA (n = 96), gout (n = 54), osteoarthritis (n = 47), healthy controls (n = 86) and synovial fluid samples from OA (n = 8) and RA (n = 8) patients, respectively. Additionally, an external validation in cohort 2 (n = 156) comprising various inflammatory diseases was employed. RESULTS The study revealed a distinctive upregulation of sSIRL-1 in the serum of RA compared to HC and other arthralgia diseases (p < 0.0001), which also displayed a significant elevation in synovial fluid from RA compared to OA (p < 0.05). Notably, sSIRL-1 levels exhibited a significant decrease in patients who achieved disease remission (p < 0.05). Furthermore, the diagnostic accuracy of RA was enhanced when sSIRL-1 was combined with anti-CCP and RF, yielding an impressive AUC value of 0.950. CONCLUSION The expression pattern of sSIRL-1 in RA, coupled with its correlation with disease activity, underscores its potential clinical utility for both diagnosis and disease monitoring in RA patients. This study offers valuable insights into the evolving diagnostic landscape of RA.
Collapse
Affiliation(s)
- Zhen Xv
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Xuejing Xv
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University, Nanjing, People's Republic of China
| | - Nianzhen Chen
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Jiayi Yuan
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Jing Li
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Lan Wang
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Shanshan Yu
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Gen Li
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Menglei Ding
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China
| | - Ming Zong
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China.
| | - Lieying Fan
- Department of Clinical Laboratory, The Affiliated East Hospital of Tongji University, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, People's Republic of China.
| |
Collapse
|
11
|
Xia J, Zhang Z, Huang Y, Wang Y, Liu G. Regulation of neutrophil extracellular traps in cancer. Int J Cancer 2024; 154:773-785. [PMID: 37815294 DOI: 10.1002/ijc.34750] [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: 05/12/2023] [Revised: 08/22/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023]
Abstract
Neutrophil extracellular trap (NET) is one of the defense functions of neutrophils, which has a rapid ability to kill infections and is also crucial in a variety of immune-associated diseases including infections, tumors and autoimmune diseases. Recent studies have shown that NETs are closely related to the development of tumors. The regulatory role of NETs in tumors has been of interest to researchers. In addition to awakening latent tumor cells, NETs can also promote the proliferation and development of tumor cells and their metastasis to other sites. At the same time, NETs also have the effect of inhibiting tumors. At present, there are some new advances in the impact of NETs on tumor development, which will provide a more theoretical basis for developing NET-targeted drugs. Therefore, this review just summarized the formation process of NETs, the regulation of tumor development and the treatment methods based on NETs.
Collapse
Affiliation(s)
- Jingxuan Xia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Zhiyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yijin Huang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
12
|
Li Y, Wu Y, Huang J, Cao X, An Q, Peng Y, Zhao Y, Luo Y. A variety of death modes of neutrophils and their role in the etiology of autoimmune diseases. Immunol Rev 2024; 321:280-299. [PMID: 37850797 DOI: 10.1111/imr.13284] [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] [Indexed: 10/19/2023]
Abstract
Neutrophils are important in the context of innate immunity and actively contribute to the progression of diverse autoimmune disorders. Distinct death mechanisms of neutrophils may exhibit specific and pivotal roles in autoimmune diseases and disease pathogenesis through the orchestration of immune homeostasis, the facilitation of autoantibody production, the induction of tissue and organ damage, and the incitement of pathological alterations. In recent years, more studies have provided in-depth examination of various neutrophil death modes, revealing nuances that challenge conventional understanding and underscoring their potential clinical utility in diagnosis and treatment. This review explores the multifaceted processes and characteristics of neutrophil death, with a focus on tailored investigations within various autoimmune diseases. It also highlights the potential interplay between neutrophil death and the landscape of autoimmune disorders. The review encapsulates the pertinent pathways implicated in various neutrophil death mechanisms across diverse autoimmune diseases while also charts possible avenues for future research.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinlan Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingang Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xue Cao
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Qiyuan An
- School of Inspection and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yun Peng
- Department of Rheumatology and Clinical Immunology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Yi Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Kuley R, Duvvuri B, Wallin JJ, Bui N, Adona MV, O’Connor NG, Sahi SK, Stanaway IB, Wurfel MM, Morrell ED, Liles WC, Bhatraju PK, Lood C. Mitochondrial N-formyl methionine peptides contribute to exaggerated neutrophil activation in patients with COVID-19. Virulence 2023; 14:2218077. [PMID: 37248708 PMCID: PMC10231045 DOI: 10.1080/21505594.2023.2218077] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 05/31/2023] Open
Abstract
Neutrophil dysregulation is well established in COVID-19. However, factors contributing to neutrophil activation in COVID-19 are not clear. We assessed if N-formyl methionine (fMet) contributes to neutrophil activation in COVID-19. Elevated levels of calprotectin, neutrophil extracellular traps (NETs) and fMet were observed in COVID-19 patients (n = 68), particularly in critically ill patients, as compared to HC (n = 19, p < 0.0001). Of note, the levels of NETs were higher in ICU patients with COVID-19 than in ICU patients without COVID-19 (p < 0.05), suggesting a prominent contribution of NETs in COVID-19. Additionally, plasma from COVID-19 patients with mild and moderate/severe symptoms induced in vitro neutrophil activation through fMet/FPR1 (formyl peptide receptor-1) dependent mechanisms (p < 0.0001). fMet levels correlated with calprotectin levels validating fMet-mediated neutrophil activation in COVID-19 patients (r = 0.60, p = 0.0007). Our data indicate that fMet is an important factor contributing to neutrophil activation in COVID-19 disease and may represent a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Center for Life Sciences, Mahindra University, Hyderabad, India
| | - Bhargavi Duvvuri
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| | | | - Nam Bui
- Biomarker Sciences, Gilead Sciences Inc, Foster City, CA, USA
| | - Mary Vic Adona
- Biomarker Sciences, Gilead Sciences Inc, Foster City, CA, USA
| | - Nicholas G. O’Connor
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Sharon K. Sahi
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Ian B. Stanaway
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Mark M. Wurfel
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Eric D. Morrell
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - W. Conrad Liles
- Department of Medicine, University of Washington, Seattle, WA, USA
- Sepsis Center of Research Excellence-UW (SCORE-UW), University of Washington, Seattle, WA, USA
| | - Pavan K. Bhatraju
- Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
- Sepsis Center of Research Excellence-UW (SCORE-UW), University of Washington, Seattle, WA, USA
| | - Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Frade-Sosa B, Sanmartí R. Neutrophils, neutrophil extracellular traps, and rheumatoid arthritis: An updated review for clinicians. REUMATOLOGIA CLINICA 2023; 19:515-526. [PMID: 37867028 DOI: 10.1016/j.reumae.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/02/2023] [Indexed: 10/24/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by the presence of autoantibodies. Research on the pathogenic mechanisms involved in systemic autoimmune diseases has largely focused on the involvement of the adaptive immune system with dysregulated responses of T and B cells. However, in recent years, there is increasing evidence of the significant role played by the innate immune system, particularly neutrophils, in these diseases, particularly in RA. Neutrophil extracellular traps (NETs) are extracellular structures composed of remodeled and concentrated chromatin with DNA, histones, and neutrophil proteins, and were first described in 2004. It has been studied that NETs may play a pathogenic role in RA and could be a source of autoantigens, increasing the immune response in the form of autoantibodies in this disease. The possible role of NETs and other markers of neutrophil activation as biomarkers of activity in RA and other immune-mediated diseases has also been studied. This article reviews the role of NETs in RA. It discusses the role of neutrophils and the latest advances in NETs, especially their involvement in autoimmune phenomena in RA. Finally, a literature review is conducted on the determination of NETs in peripheral blood and their relationship as a biomarker of RA activity, as well as their potential role in disease monitoring.
Collapse
Affiliation(s)
- Beatriz Frade-Sosa
- Servicio de Reumatología, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Raimon Sanmartí
- Servicio de Reumatología, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Hammer HB, Pedersen SL, Jonsson MK, Mathsson-Alm L, Gehring I, Sexton J, Haavardsholm EA, Askling J. Calprotectin, a sensitive marker of inflammation, is robustly assessed in plasma from patients with early or established rheumatoid arthritis by use of different laboratory methods. Scand J Clin Lab Invest 2023; 83:330-335. [PMID: 37343245 DOI: 10.1080/00365513.2023.2225223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/01/2023] [Accepted: 06/11/2023] [Indexed: 06/23/2023]
Abstract
Calprotectin (S100A8/S100A9, MRP8/MRP14) is a major leukocyte protein found to be more sensitive than C-Reactive Protein (CRP) and Erythrocyte Sedimentation Rate (ESR) as a marker of inflammation in patients with rheumatoid arthritis (RA). The present objective was to explore the robustness of calprotectin assessments by comparing two different laboratory methods assessing calprotectin in plasma samples from patients with early or established RA. A total of 212 patients with early RA (mean (SD) age 52(13.3) years, disease duration 0.6(0.5) years) and 177 patients with established RA (mean (SD) age 52.9(13.0) years, disease duration 10.0(8.8) years) were assessed by clinical, laboratory, and ultrasound examinations. Frozen plasma samples (-80 °C) were analysed for calprotectin levels at baseline, 1, 2, 3, 6 and 12 months by use of either enzyme-linked immunosorbent assay (ELISA) or fluoroenzyme immunoassay (FEIA). The ELISA technique used kits from Calpro AS and the FEIA technology was assessed on an automated Thermo Fisher Scientific instrument. The results showed high correlations between the two methods at baseline and during follow-up, with Spearman correlation at baseline 0.93 (p < 0.001) in the early and 0.96 (p < 0.001) in the established RA cohorts. The correlations between each of the two calprotectin assessments and clinical examinations had similar range. Calprotectin correlated well with clinical examinations, with at least as high correlations as CRP and ESR. The present study showed similar results for the two analytical methods, supporting the robustness of calprotectin analyses, and suggest calprotectin in plasma to be included in the assessments offered by clinical routine laboratories.
Collapse
Affiliation(s)
- Hilde Berner Hammer
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Maria K Jonsson
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | | | | | - Joe Sexton
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Espen A Haavardsholm
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Johan Askling
- Department of Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
16
|
Michailidou D, Johansson L, Kuley R, Wang T, Hermanson P, Rantapää-Dahlqvist S, Lood C. Immune complex-mediated neutrophil activation in patients with polymyalgia rheumatica. Rheumatology (Oxford) 2023; 62:2880-2886. [PMID: 36562570 PMCID: PMC10393430 DOI: 10.1093/rheumatology/keac722] [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: 09/29/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Neutrophils are important in host defence. However, neutrophils are also linked to inflammation and organ damage. The purpose of this study was to assess whether markers of neutrophil activation are increased in PMR. METHODS Levels of immune complexes (IC), calprotectin and neutrophil extracellular traps (NETs) were measured in plasma of healthy individuals (n = 30) and patients with PMR (n = 60), at flare and upon treatment with glucocorticoids using ELISA. Plasma-mediated neutrophil activation was assessed in presence of an FcγRIIA inhibitory antibody (IV.3). RESULTS Plasma levels of calprotectin and NETs were elevated in PMR (P < 0.001). Mechanistically, neutrophil activation was driven by ICs, present in plasma, able to up-regulate neutrophil activation markers CD66b and CD11b (P < 0.0001) in an FcγRIIA-dependent manner (P < 0.01). Of note, circulating levels of IC correlated with plasma induced CD66b and CD11b (r = 0.51, P = 0.004, and r = 0.46, P = 0.01, respectively) and decreased after glucocorticoid therapy. In contrast to NETs, calprotectin significantly decreased after glucocorticoid therapy (P < 0.001) and was higher in PMR without overlapping GCA compared with patients with overlapping disease (P = 0.014). Interestingly, musculoskeletal involvement was associated with elevated levels of calprotectin before initiation of glucocorticoid therapy (P = 0.036). CONCLUSIONS Neutrophil activation, including NET formation, is increased in PMR, through IC-mediated engagement of FcγRIIA. Clinically, neutrophil activation is associated with musculoskeletal involvement, with calprotectin, but not NETs, being a biomarker of treatment response in PMR patients. In all, IC-mediated neutrophil activation is a central process in PMR pathogenesis identifying potential novel therapeutic targets (FcγRIIA), as well as soluble markers for disease monitoring (calprotectin).
Collapse
Affiliation(s)
- Despina Michailidou
- Division of Rheumatology, Department of Medicine,University of Washington, Seattle, WA, USA
| | - Linda Johansson
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | - Runa Kuley
- Division of Rheumatology, Department of Medicine,University of Washington, Seattle, WA, USA
- Center of Life Sciences, Mahindra University, Hyderabad, India
| | - Ting Wang
- Division of Rheumatology, Department of Medicine,University of Washington, Seattle, WA, USA
| | - Payton Hermanson
- Division of Rheumatology, Department of Medicine,University of Washington, Seattle, WA, USA
| | | | - Christian Lood
- Division of Rheumatology, Department of Medicine,University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Heshin-Bekenstein M, Baron S, Schulert G, Shusterman A, Fidel V, Ben-Shahar Y, Shukrun R, Binenbaum Y, Elhasid R. Neutrophils extracellular traps formation may serve as a biomarker for disease activity in oligoarticular juvenile idiopathic arthritis: a pilot study. Arthritis Res Ther 2023; 25:135. [PMID: 37525216 PMCID: PMC10388488 DOI: 10.1186/s13075-023-03104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease in children, causing significant morbidity. Despite the dramatic improvement in treatment, many patients do not achieve complete remission, and biomarkers for subclinical disease, flares, and response to treatment are lacking. Neutrophils and neutrophil extracellular traps (NETs) play key roles in the pathogenesis of autoimmune and inflammatory conditions. In this study, we characterized neutrophil enzyme activity and NETs formation in oligoarticular and polyarticular JIA and explored their association with disease activity. METHODS Neutrophils from 6 healthy controls and 7 patients with oligoarticular and polyarticular JIA were freshly isolated at time of diagnosis and after glucocorticoid intra-articular injection. Enzymatic activity of neutrophil granular enzymes was monitored by colorimetry and PMA-activated NETs formation was assessed using fluorescent microscopy. RESULTS In this pilot and feasibility study, we revealed that NETs were significantly increased in oligoarticular JIA patients at time of diagnosis compared to healthy controls. Anti-inflammatory treatment using intra-articular steroid injection normalized NETs formation in these patients. Correlation between NETs formation and clinical Juvenile Activity Disease Activity Score-10 (cJADAS-10) was linear and significant (P = 0.007) in oligo but not in poly JIA patients. CONCLUSIONS This is the first study exploring the link of NETs formation with oligo and poly JIA activity. We demonstrated a statistically significant linear correlation between cJADAS-10 and NETs formation in oligo but not in poly JIA patients. Hence, we suggest that NETs may reflect clinical disease activity in JIA, and may serve as a putative biomarker. Further work is needed to validate these initial results and determine the dynamics of NETs formation in JIA.
Collapse
Affiliation(s)
- Merav Heshin-Bekenstein
- Pediatric Rheumatology Service, Dana Dwek Children's Hospital of Tel Aviv Medical Center, Tel Aviv, Israel.
- School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Szilvia Baron
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Grant Schulert
- Division of Rheumatology, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Anna Shusterman
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Victoria Fidel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yoav Ben-Shahar
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Pediatric Surgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Rachel Shukrun
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yoav Binenbaum
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ronit Elhasid
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hemato-Oncology Research Laboratory, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Pediatric Hemato-Oncology, Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
18
|
Moore RE, Wang T, Duvvuri B, Feser ML, Deane KD, Solomon JJ, Lee Nelson J, Demoruelle MK, Lood C. Prediction of Erosive Disease Development by Antimitochondrial Antibodies in Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:890-899. [PMID: 36580020 PMCID: PMC10238559 DOI: 10.1002/art.42428] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Mitochondria are found in the extracellular space in rheumatoid arthritis (RA). However, whether mitochondria are a source of autoantigens in RA has not been carefully addressed. Thus, we undertook this study to investigate the presence and significance of antimitochondrial antibodies (AMAs) in patients with RA. METHODS AMAs were measured in serum samples from 3 cross-sectional cohorts of RA patients (n = 95, n = 192, and n = 117) and healthy individuals (n = 38, n = 72, and n = 50) using a flow cytometry-based assay. Further, AMAs were detected using an anti-mitofusin-1 (anti-MFN-1) IgG enzyme-linked immunosorbent assay and Western blot analysis. A longitudinal inception cohort, followed up for a median of 8 years, was used to study disease progression. RESULTS AMA levels were elevated in RA patients from all 3 cohorts as compared to healthy individuals (P < 0.001, P < 0.05, and P < 0.01), with a range of 14-26% positivity. Levels of anti-MFN-1 antibodies correlated with AMA levels (r = 0.31, P = 0.006) and were elevated in RA patients as compared to healthy individuals (P < 0.001). The presence of AMAs was associated with erosive disease (P < 0.05) and interstitial lung disease (P < 0.01). Further, AMA levels were found to predict erosive disease (odds ratio [OR] 4.59, P = 0.006) and joint space narrowing (OR 3.08, P = 0.02) independent of anti-citrullinated protein antibodies. Finally, anti-MFN-1 antibodies identified seronegative patients developing erosive disease (OR 9.33; P = 0.02). CONCLUSION Our findings demonstrate the presence of novel autoantibodies targeting mitochondria in the setting of RA. AMAs were used to stratify patients based on disease phenotype and to predict development of erosive disease, including in patients with seronegative disease. Our results highlight the essential role of mitochondria in the pathogenesis of RA and suggest a possible benefit of therapies targeting mitochondrial-mediated inflammation and clearance in these patients.
Collapse
Affiliation(s)
- Richard E. Moore
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Ting Wang
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Marie L. Feser
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | - Joshua J. Solomon
- Center for Interstitial Lung Disease, National Jewish Health, Denver, CO, USA
| | - J. Lee Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Frade-Sosa B, Chacur CA, Augé JM, Ponce A, Sarmiento-Monroy JC, Azuaga AB, Sapena N, Ramírez J, Ruiz-Esquide V, Morlà R, Farietta S, Corzo P, Cañete JD, Sanmartí R, Gómez-Puerta JA. Calprotectin in Patients with Rheumatic Immunomediated Adverse Effects Induced by Checkpoints Inhibitors. Cancers (Basel) 2023; 15:cancers15112984. [PMID: 37296947 DOI: 10.3390/cancers15112984] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND this is an exploratory study to evaluate calprotectin serum levels in patients with rheumatic immune-related adverse events (irAEs) induced by immune checkpoint inhibitor (ICI) treatment. METHODS this is a retrospective observational study including patients with irAEs rheumatic syndromes. We compared the calprotectin levels to those in a control group of patients with RA and with a control group of healthy individuals. Additionally, we included a control group of patients treated with ICI but without irAEs to check calprotectin levels. We also analysed the performance of calprotectin for the identification of active rheumatic disease using receiver operating characteristic curves (ROC). RESULTS 18 patients with rheumatic irAEs were compared to a control group of 128 RA patients and another group of 29 healthy donors. The mean calprotectin level in the irAE group was 5.15 μg/mL, which was higher than the levels in both the RA group (3.19 μg/mL) and the healthy group (3.81 μg/mL) (cut-off 2 μg/mL). Additionally, 8 oncology patients without irAEs were included. In this group, calprotectin levels were similar to those of the healthy controls. In patients with active inflammation, the calprotectin levels in the irAE group were significantly higher (8.43 μg/mL) compared to the RA group (3.94 μg/mL). ROC curve analysis showed that calprotectin had a very good discriminatory capacity to identify inflammatory activity in patients with rheumatic irAEs (AUC of 0.864). CONCLUSIONS the results suggest that calprotectin may serve as a marker of inflammatory activity in patients with rheumatic irAEs induced by treatment with ICIs.
Collapse
Affiliation(s)
- Beatriz Frade-Sosa
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Chafik Alejandro Chacur
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Josep M Augé
- Department of Biochemistry and Molecular Genetics (CDB), Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Andrés Ponce
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Juan C Sarmiento-Monroy
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ana Belén Azuaga
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Nuria Sapena
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Julio Ramírez
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Virginia Ruiz-Esquide
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Morlà
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Sandra Farietta
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Patricia Corzo
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Juan D Cañete
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Raimon Sanmartí
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - José A Gómez-Puerta
- Department of Rheumatology, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
20
|
Heidt C, Kämmerer U, Fobker M, Rüffer A, Marquardt T, Reuss-Borst M. Assessment of Intestinal Permeability and Inflammation Bio-Markers in Patients with Rheumatoid Arthritis. Nutrients 2023; 15:nu15102386. [PMID: 37242269 DOI: 10.3390/nu15102386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Increased intestinal permeability and inflammation, both fueled by dysbiosis, appear to contribute to rheumatoid arthritis (RA) pathogenesis. This single-center pilot study aimed to investigate zonulin, a marker of intestinal permeability, and calprotectin, a marker of intestinal inflammation, measured in serum and fecal samples of RA patients using commercially available kits. We also analyzed plasma lipopolysaccharide (LPS) levels, a marker of intestinal permeability and inflammation. Furthermore, univariate, and multivariate regression analyses were carried out to determine whether or not there were associations of zonulin and calprotectin with LPS, BMI, gender, age, RA-specific parameters, fiber intake, and short-chain fatty acids in the gut. Serum zonulin levels were more likely to be abnormal with a longer disease duration and fecal zonulin levels were inversely associated with age. A strong association between fecal and serum calprotectin and between fecal calprotectin and LPS were found in males, but not in females, independent of other biomarkers, suggesting that fecal calprotectin may be a more specific biomarker than serum calprotectin is of intestinal inflammation in RA. Since this was a proof-of-principle study without a healthy control group, further research is needed to validate fecal and serum zonulin as valid biomarkers of RA in comparison with other promising biomarkers.
Collapse
Affiliation(s)
- Christina Heidt
- University of Muenster, D-48149 Muenster, Germany
- Department of General Pediatrics, Metabolic Diseases, University of Muenster, Albert-Schweitzer-Campus, D-48149 Muenster, Germany
| | - Ulrike Kämmerer
- Department of Obstetrics and Gynaecology, University Hospital of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Manfred Fobker
- Centre of Laboratory Medicine, University Hospital Muenster, D-48149 Muenster, Germany
| | | | - Thorsten Marquardt
- Department of General Pediatrics, Metabolic Diseases, University of Muenster, Albert-Schweitzer-Campus, D-48149 Muenster, Germany
| | - Monika Reuss-Borst
- Hescuro Center for Rehabilitation and Prevention Bad Bocklet, D-97708 Bad Bocklet, Germany
- Department of Nephrology and Rheumatology, Georg-August University of Goettingen, D-37075 Goettingen, Germany
| |
Collapse
|
21
|
Lantieri MA, Perdomo Trejo JR, Le Q, Dighe A, Cui Q, Yang X. Formyl peptide receptors in bone research. Connect Tissue Res 2023; 64:229-237. [PMID: 36440821 PMCID: PMC10164673 DOI: 10.1080/03008207.2022.2149397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE/AIM OF THE STUDY The formyl peptide receptor (FPR) participates in the immune response, with roles in infection and inflammation. In this review article, we summarize the current literature on these roles before discussing the function of FPRs in the pathogenesis of musculoskeletal disorders including osteoarthritis (OA), degenerative disc disease (DDD), and rheumatoid arthritis (RA). Additionally, we discuss the potential diagnostic and therapeutic roles of FPRs in these domains. METHODS PubMed and Ovid MEDLINE searches were performed from 1965 through March 2022. Keywords included "FPR, tissue expression, inflammation, infection, musculoskeletal disorder, bone, rheumatoid arthritis, osteoarthritis, degenerative disc disease, mitochondria." RESULTS Sixty-nine studies were included in this review article. FPRs appear to be ubiquitous in the pathogenesis, diagnosis, and treatment of common musculoskeletal disorders. They can potentially be utilized for the earlier diagnosis of OA and DDD. They may be employed with mesenchymal stem cells (MSCs) to reverse OA and DDD pathologies. With anti-inflammatory, anti-osteolytic, and pro-angiogenic functions, they may broaden treatment options in RA. CONCLUSIONS FPRs appear to be heavily involved in the pathogenesis of common musculoskeletal conditions, including arthritis, degenerative disc disease, and rheumatoid arthritis. Furthermore, they demonstrate much promise in the diagnosis and treatment of these conditions. Their roles should continue to be explored.
Collapse
Affiliation(s)
- Mark A. Lantieri
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA
| | | | - Quang Le
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA
| | - Abhijit Dighe
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA
| | - Quanjun Cui
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA
| | - Xinlin Yang
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA
| |
Collapse
|
22
|
Michailidou D, Kuley R, Wang T, Hermanson P, Grayson PC, Cuthbertson D, Khalidi NA, Koening CL, Langford CA, McAlear CA, Moreland LW, Pagnoux C, Seo P, Specks U, Sreih AG, Warrington KJ, Monach PA, Merkel PA, Lood C. Neutrophil extracellular trap formation in anti-neutrophil cytoplasmic antibody-associated and large-vessel vasculitis. Clin Immunol 2023; 249:109274. [PMID: 36878421 PMCID: PMC10066833 DOI: 10.1016/j.clim.2023.109274] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/07/2023]
Abstract
Levels of neutrophil extracellular traps (NETs) were measured in plasma of healthy controls (HC, n = 30) and patients with granulomatosis with polyangiitis (GPA, n = 123), microscopic polyangiitis (MPA, n = 61), Takayasu's arteritis (TAK, n = 58), and giant cell arteritis (GCA, n = 68), at times of remission or activity and correlated with levels of the platelet-derived thrombospondin-1 (TSP-1). Levels of NETs were elevated during active disease in patients with GPA (p < 0.0001), MPA (p = 0.0038), TAK (p < 0.0001), and GCA (p < 0.0001), and in remission for GPA, p < 0.0001, MPA, p = 0.005, TAK, p = 0.03, and GCA, p = 0.0009. All cohorts demonstrated impaired NET degradation. Patients with GPA (p = 0.0045) and MPA (p = 0.005) had anti-NET IgG antibodies. Patients with TAK had anti-histone antibodies (p < 0.01), correlating with presence of NETs. Levels of TSP-1 were increased in all patients with vasculitis, and associated with NET formation. NET formation is a common process in vasculitides. Targeting NET formation or degradation could be potential therapeutic approaches for vasculitides.
Collapse
Affiliation(s)
| | - Runa Kuley
- Division of Rheumatology, University of Washington, Seattle, USA; Center for Life Sciences, Mahindra University, Hyderabad, India
| | - Ting Wang
- Division of Rheumatology, University of Washington, Seattle, USA
| | - Payton Hermanson
- Division of Rheumatology, University of Washington, Seattle, USA
| | - Peter C Grayson
- Systemic Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - David Cuthbertson
- Health Informatics Institute, University of South Florida, South Florida, FL, USA
| | - Nader A Khalidi
- Division of Rheumatology, Mc Master University, Ontario, Canada
| | | | | | - Carol A McAlear
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | - Larry W Moreland
- Division of Rheumatology and Clinical Immunology, University of Colorado, Denver, CO, USA
| | | | - Philip Seo
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Antoine G Sreih
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Paul A Monach
- Division of Rheumatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter A Merkel
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, USA.
| |
Collapse
|
23
|
Selldén T, Mårdh CK, Joelsson M, Vikgren J, Johnsson Å, Larsson G, Glinatsi D, Stubendorff K, Svensson K, Gjertsson I, Rudin A, Lundell AC, Ekwall AKH. Radiographic airway abnormalities in untreated early rheumatoid arthritis are associated with peripheral neutrophil activation. Arthritis Res Ther 2023; 25:44. [PMID: 36941690 PMCID: PMC10026468 DOI: 10.1186/s13075-023-03019-5] [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: 12/09/2022] [Accepted: 02/22/2023] [Indexed: 03/22/2023] Open
Abstract
BACKGROUND The role of the lung for the initiation and progression of rheumatoid arthritis (RA) is still unclear. Up to 10% of RA patients develop interstitial lung disease which remains a clinical challenge. Understanding early disease mechanisms is of great importance. The objective of this study was to determine whether there is an association between peripheral neutrophil phenotypes and presence of pulmonary abnormalities (PA) on chest high-resolution computed tomography (HRCT) in untreated early RA (ueRA). METHODS Clinical data and blood were collected, and HRCT performed at diagnosis on 30 consecutive anti-citrullinated protein antibody (ACPA) and/or rheumatoid factor (RF) positive ueRA patients. HRCTs were evaluated for the presence of RA-associated parenchymal, airway and/or pleural abnormalities. Expression of phenotype markers on neutrophils were determined by flow cytometry. Levels of calprotectin, ACPA and RF were measured using immunoassays. RESULTS The frequency of having any PA was 60%. Airway abnormalities were present in 50%, parenchymal nodules in 43% and interstitial lung abnormalities (ILA) in 10%. Unsupervised multivariate data analysis showed clustering of any PA with neutrophil activation, parameters of inflammation and RF titres. In univariate analysis, the patients with PA displayed significantly increased CD11b and decreased CD62L expression on neutrophils (1.2-fold, p = 0.014; 0.8-fold, p = 0.012) indicating activation and significantly increased RF IgM titre and CRP (5.7-fold, p = 0.0025; 2.3-fold, p = 0.0035) as compared to no PA. Titres of RF, but not ACPA, correlated with expression of the neutrophil activation marker CD11b. A stratified analysis demonstrated that airway involvement was the PA subtype with the strongest association with neutrophil activation. CONCLUSION We report a strong association between radiographic airway findings and activation of circulating neutrophils in early RA supporting a role of innate immunity and the lung at disease onset. Our results also indicate different contributions of RF and ACPA in the RA pathogenesis.
Collapse
Grants
- ALFGBG-942903, 943046 the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement
- ALFGBG-942903, 943046 the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement
- ALFGBG-942903, 943046 the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement
- ALFGBG-942903, 943046 the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement
- ALFGBG-942903, 943046 the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement
- R-940577 Swedish Rheumatism Association
- R-940577 Swedish Rheumatism Association
- R-940577 Swedish Rheumatism Association
- GLS-935160, 961518 Gothenburg Society of Medicine
- GLS-935160, 961518 Gothenburg Society of Medicine
- GLS-935160, 961518 Gothenburg Society of Medicine
- GLS-935160, 961518 Gothenburg Society of Medicine
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- VGFOUREG-932185, 940809 Local Research and Development Council in Västra Götaland Region, Sweden
- 2020-06193 Swedish Research Council
- 2020-06193 Swedish Research Council
- 2020-06193 Swedish Research Council
- 2020-06193 Swedish Research Council
- The Rune and Ulla Amlöv foundation
- University of Gothenburg
Collapse
Affiliation(s)
- Tilia Selldén
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
| | - Carina Kärrman Mårdh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin Joelsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jenny Vikgren
- Department of Radiology, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Åse Johnsson
- Department of Radiology, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gunilla Larsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
| | - Daniel Glinatsi
- Department of Rheumatology, Skaraborg Hospital, Skövde, Sweden
| | | | - Karin Svensson
- Department of Rheumatology, Skaraborg Hospital, Skövde, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna-Carin Lundell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Guldhedsgatan 10A, Gothenburg, SE-405 30, Sweden.
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
24
|
Shahidi M, Amirzargar MR. The impact of neutrophil extracellular traps in coronavirus disease - 2019 pathophysiology. Blood Coagul Fibrinolysis 2023; 34:87-92. [PMID: 36719805 DOI: 10.1097/mbc.0000000000001181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by novel coronavirus-2019 (nCoV-2019), is a highly contagious disease with high mortality and morbidity risk. Infected people may suffer from respiratory infections, which may be more progressive in patients with a defective immune system and underlying medical problems. In this regard, the cells involved in the innate immune system, play a decisive role in disease progression and complication development. Pathogen entrapment is the critical role of neutrophil extracellular traps (NETosis). This process involves the widespread release of fibrous structures by the stimulant-activated neutrophils. These fibrous structures are composed of cytosolic proteins and granular contents brought together by a network of released chromatins. This network can inhibit the spread of pathogens by their entrapment. Moreover, NETosis damage the host by producing toxic agents and triggering thrombosis. Therefore, this phenomenon may act as a double-edged sword. Regarding the rapid expansion of COVID-19, it is crucial to examine the involvement of NETosis in infected patients. This study aims to discuss NETosis participation to show its probable association with increased risk of thrombogenicity and help develop new therapeutic approaches in the battle against this viral disease.
Collapse
Affiliation(s)
- Minoo Shahidi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
25
|
Wareing N, Mohan V, Taherian R, Volkmann ER, Lyons MA, Wilhalme H, Roth MD, Estrada-y-Martin RM, Skaug B, Mayes MD, Tashkin DP, Assassi S. Blood Neutrophil Count and Neutrophil-to-Lymphocyte Ratio for Prediction of Disease Progression and Mortality in Two Independent Systemic Sclerosis Cohorts. Arthritis Care Res (Hoboken) 2023; 75:648-656. [PMID: 35287250 PMCID: PMC9470772 DOI: 10.1002/acr.24880] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/16/2022] [Accepted: 03/08/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To assess the predictive significance of blood neutrophil count and the ratio between neutrophil and lymphocyte count (neutrophil-to-lymphocyte ratio [NLR]) for disease severity and mortality in systemic sclerosis (SSc). METHODS Neutrophil and lymphocyte counts were prospectively measured in the Genetics versus Environment in Scleroderma Outcome Study (GENISOS) and the Scleroderma Lung Study II (SLS II). Forced vital capacity percent predicted (FVC%) and modified Rodnan skin thickness score (MRSS) were used as surrogate measures for disease severity. Longitudinal analyses were performed using generalized linear mixed models. Cox proportional hazards models evaluated the predictive significance of these cell counts for mortality. RESULTS Of the 447 SSc patients in the GENISOS cohort at the time of analysis, 377 (84.3%) had available baseline blood neutrophil and lymphocyte counts. Higher baseline neutrophil count and NLR predicted lower serially obtained FVC% (b = -4.74, P = 0.009 and b = -2.68, P = 0.028, respectively) and higher serially obtained MRSS (b = 4.07, P < 0.001 and b = 2.32, P < 0.001, respectively). Longitudinal neutrophil and NLR measurements also significantly correlated with lower concurrently obtained FVC% measurements and higher concurrently obtained MRSS. Baseline neutrophil count and NLR predicted increased risk of long-term mortality, even after adjustment for baseline demographic and clinical factors (hazard ratio [HR] 1.42, P = 0.02 and HR 1.48, P < 0.001, respectively). The predictive significance of higher baseline neutrophil count and NLR for declining FVC% and increased long-term mortality was confirmed in the SLS II. CONCLUSION Higher blood neutrophil count and NLR are predictive of more severe disease course and increased mortality, indicating that these easily obtainable laboratory studies might be a reflection of pathologic immune processes in SSc.
Collapse
Affiliation(s)
- Nancy Wareing
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | - Vishnu Mohan
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | - Rana Taherian
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | | | - Marka A. Lyons
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | | | | | | | - Brian Skaug
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | - Maureen D. Mayes
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| | | | - Shervin Assassi
- McGovern Medical School at University of Texas Health Science Center at Houston, TX, USA
| |
Collapse
|
26
|
Cai Y, Liang J, Chen X, Zhang G, Jing Z, Zhang R, Lv L, Zhang W, Dang X. Synovial fluid neutrophil extracellular traps could improve the diagnosis of periprosthetic joint infection. Bone Joint Res 2023; 12:113-120. [PMID: 36718647 PMCID: PMC9950667 DOI: 10.1302/2046-3758.122.bjr-2022-0391.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
AIMS This study aimed to explore the diagnostic value of synovial fluid neutrophil extracellular traps (SF-NETs) in periprosthetic joint infection (PJI) diagnosis, and compare it with that of microbial culture, serum ESR and CRP, synovial white blood cell (WBC) count, and polymorphonuclear neutrophil percentage (PMN%). METHODS In a single health centre, patients with suspected PJI were enrolled from January 2013 to December 2021. The inclusion criteria were: 1) patients who were suspected to have PJI; 2) patients with complete medical records; and 3) patients from whom sufficient synovial fluid was obtained for microbial culture and NET test. Patients who received revision surgeries due to aseptic failure (AF) were selected as controls. Synovial fluid was collected for microbial culture and SF-WBC, SF-PNM%, and SF-NET detection. The receiver operating characteristic curve (ROC) of synovial NET, WBC, PMN%, and area under the curve (AUC) were obtained; the diagnostic efficacies of these diagnostic indexes were calculated and compared. RESULTS The levels of SF-NETs in the PJI group were significantly higher than those of the AF group. The AUC of SF-NET was 0.971 (95% confidence interval (CI) 0.903 to 0.996), the sensitivity was 93.48% (95% CI 82.10% to 98.63%), the specificity was 96.43% (95% CI 81.65% to 99.91%), the accuracy was 94.60% (95% CI 86.73% to 98.50%), the positive predictive value was 97.73%, and the negative predictive value was 90%. Further analysis showed that SF-NET could improve the diagnosis of culture-negative PJI, patients with PJI who received antibiotic treatment preoperatively, and fungal PJI. CONCLUSION SF-NET is a novel and ideal synovial fluid biomarker for PJI diagnosis, which could improve PJI diagnosis greatly.Cite this article: Bone Joint Res 2023;12(2):113-120.
Collapse
Affiliation(s)
- Yuanqing Cai
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China,Department of Orthopaedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jialin Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Xiaoqing Chen
- Department of Orthopaedics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Guangyang Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Zhaopu Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Rupeng Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Leifeng Lv
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China
| | - Wenming Zhang
- Department of Orthopaedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoqian Dang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an, China, Xiaoqian Dang. E-mail:
| |
Collapse
|
27
|
van der Linden M, Kumari S, Montizaan D, van Dalen S, Kip A, Foster M, Reinieren-Beeren I, Neubert E, Erpenbeck L, Waaijenberg K, Bruurmijn T, Te Poele R, van Zandvoort P, Vink P, Meldrum E, van Es H, Chirivi RGS. Anti-citrullinated histone monoclonal antibody CIT-013, a dual action therapeutic for neutrophil extracellular trap-associated autoimmune diseases. MAbs 2023; 15:2281763. [PMID: 38031350 DOI: 10.1080/19420862.2023.2281763] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Neutrophil extracellular traps (NETs) contribute to the pathophysiology of multiple inflammatory and autoimmune diseases. Targeting the NETosis pathway has demonstrated significant therapeutic potency in various disease models. Here, we describe a first-in-class monoclonal antibody (CIT-013) with high affinity for citrullinated histones H2A and H4, which inhibits NETosis and reduces tissue NET burden in vivo with significant anti-inflammatory consequences. We provide a detailed understanding of the epitope selectivity of CIT-013. Detection of CIT-013 epitopes in rheumatoid arthritis (RA) synovium provides evidence that RA is an autoimmune disease with excessive citrullinated NETs that can be targeted by CIT-013. We show that CIT-013 acts upon the final stage of NETosis, binding to its chromatin epitopes when plasma membrane integrity is compromised to prevent NET release. Bivalency of CIT-013 is necessary for NETosis inhibition. In addition, we show that CIT-013 binding to NETs and netting neutrophils enhance their phagocytosis by macrophages in an Fc-dependent manner. This is confirmed using a murine neutrophilic airway inflammation model where a mouse variant of CIT-013 reduced tissue NET burden with significant anti-inflammatory consequences. CIT-013's therapeutic activity provides new insights for the development of NET antagonists and indicates the importance of a new emerging therapy for NET-driven diseases with unmet therapeutic needs.
Collapse
Affiliation(s)
| | | | | | | | - Annemarie Kip
- Research and Development, Citryll B.V, Oss, The Netherlands
| | - Martyn Foster
- Pathology, Experimental Pathology Consultancy, Benfleet, Essex, UK
| | | | - Elsa Neubert
- Department of Dermatology, Venereology and Allergology, University Medical Center, Göttingen University, Göttingen, Germany
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Luise Erpenbeck
- Department of Dermatology, Venereology and Allergology, University Medical Center, Göttingen University, Göttingen, Germany
- Department of General Dermatology and Venereology, Clinic of Skin Diseases, University Medical Center Münster, Münster, Germany
| | | | | | - Rezie Te Poele
- Research and Development, Citryll B.V, Oss, The Netherlands
| | | | - Paul Vink
- Research and Development, Citryll B.V, Oss, The Netherlands
| | - Eric Meldrum
- Research and Development, Citryll B.V, Oss, The Netherlands
| | - Helmuth van Es
- Research and Development, Citryll B.V, Oss, The Netherlands
| | | |
Collapse
|
28
|
Pan W, Xin Q, Xu J, He J, Chen Z, Hu X, Li T, Zhu Y, Wei W, Wu Y. IgD enhances the release of neutrophil extracellular traps (NETs) via FcδR in rheumatoid arthritis patients. Int Immunopharmacol 2023; 114:109484. [PMID: 36450207 DOI: 10.1016/j.intimp.2022.109484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune inflammatory disorder affecting primarily the joints. Neutrophils and the release of neutrophil extracellular traps (NETs) contribute to the pathogenesis of RA. However, IgD, which was abnormally higher in RA, has not been studied for its pathological role in neutrophil activation and NETs formation. To investigate the effects of IgD on neutrophil activation and NETs formation via IgD receptor (FcδR), we collect peripheral blood of RA patients and established adjuvant-induced arthritis (AA) rat model. We found that the expression of FcδR on neutrophils was significantly higher in RA patients compared with healthy controls. As a specific marker of NETs, the level of citrullinated histone H3 was positively correlated with sIgD and FcδR in RA patients. IgD enhances the release of NETs and promotes the proliferation of fibroblast-like synoviocytes (FLS) from RA patients by activating neutrophils. As a competitive FcδR blocker, IgD-Fc-Ig fusion protein could significantly reduce NETs formation and FcδR expression on neutrophils in vitro. In vivo, IgD-Fc-Ig could restrain IgD-induced neutrophil activation and NETs formation, thus inhibited FLS proliferation in AA rats. Data presented here demonstrate that neutrophils could be triggered by IgD to release NETs and take part in FLS proliferation in RA patients with excessive IgD. Blocking IgD-FcδR could inhibit neutrophil activation and NETs formation, and represent an additional attractive novel therapeutic strategy for the treatment of RA.
Collapse
Affiliation(s)
- Wenwen Pan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Qianling Xin
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Jing Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Jingjing He
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Zhaoying Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Xiaoxi Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Tao Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanqing Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China.
| | - Yujing Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Provincial Institute of Translational Medicine, Hefei, China.
| |
Collapse
|
29
|
Tsioulos G, Kounatidis D, Vallianou NG, Koufopoulos N, Katsimbri P, Antoniadou A. Severe eosinophilic granulomatosis with polyangiitis responding to a combination of rituximab and mepolizumab. Am J Med Sci 2023; 365:93-98. [PMID: 35970248 DOI: 10.1016/j.amjms.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/07/2022] [Accepted: 07/21/2022] [Indexed: 01/04/2023]
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA), formerly known as Churg-Strauss Syndrome, is a multisystem antineutrophil cytoplasmic antibody (ANCA) positive vasculitis, characterized by the presence of chronic rhinosinusitis, asthma and prominent peripheral blood eosinophilia. Although the most commonly involved organ is the lung, followed by the skin, EGPA can affect any organ system. Herein, we present the complicated case of an 18-year-old male patient with severe life-threatening EGPA, with central nervous system, cardiac and gasterointestinal involvement, which was resistant to initial treatment with glucocorticoids and cyclophosphamide. The patient responded well, achieving complete remission after the addition of rituximab and mepolizumab to glucocorticoids and cyclophosphamide.
Collapse
Affiliation(s)
- Georgios Tsioulos
- Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | - Dimitris Kounatidis
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | - Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece.
| | | | - Pelagia Katsimbri
- Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | | |
Collapse
|
30
|
Korolkova AA, Khizha VV, Kozlova DI, Vasiliev DS, Maslyansky AL, Vavilova TV, Ballyuzek MF, Rybakov AV, Shevaldina ME, Yurieva KA. 14-3-3η Cytokine as a New Biomarker to Assess Rheumatoid Arthritis Disease Activity. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s002209302301012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
31
|
Gilbert BTP, Lamacchia C, Amend L, Strowig T, Rodriguez E, Palmer G, Finckh A. Brief report: Assessment of mucosal barrier integrity using serological biomarkers in preclinical stages of rheumatoid arthritis. Front Immunol 2023; 14:1117742. [PMID: 36875067 PMCID: PMC9977794 DOI: 10.3389/fimmu.2023.1117742] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Background The pathogenesis of rheumatoid arthritis (RA) is believed to initiate at mucosal sites. The so-called 'mucosal origin hypothesis of RA' postulates an increased intestinal permeability before disease onset. Several biomarkers, including lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP), have been proposed to reflect gut mucosa permeability and integrity, while serum calprotectin is a new inflammation marker proposed in RA. Methods We analyzed serum samples of individuals genetically at increased risk of RA in a nested-case-control study. Participants from a longitudinal cohort of first-degree relatives of RA patients (SCREEN-RA cohort) were divided into three pre-clinical stages of RA, based on the presence of risk factors for subsequent RA onset: 1) low-risk healthy asymptomatic controls; 2) intermediate-risk individuals without symptoms, but with RA-associated auto-immunity; 3) high-risk individuals with clinically suspect arthralgias. Five patients with newly diagnosed RA were also sampled. Serum LBP, I-FABP and calprotectin were measured using commercially available ELISA kits. Results We included 180 individuals genetically at increased risk for RA: 84 asymptomatic controls, 53 individuals with RA-associated autoimmunity and 38 high risk individuals. Serum LBP, I-FAPB or calprotectin concentrations did not differ between individuals in different pre-clinical stages of RA. Conclusion Based on the serum biomarkers LBP, I-FABP and calprotectin, we could not detect any evidence for intestinal injury in pre-clinical stages of RA.
Collapse
Affiliation(s)
- Benoît Thomas P Gilbert
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Céline Lamacchia
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence Resolving Infection Susceptibility (RESIST) (EXC 2155), Hannover Medical School, Hannover, Germany.,Center for Individualized Infection Medicine (CiiM), Hannover, Germany
| | - Emiliana Rodriguez
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Axel Finckh
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
32
|
Inciarte-Mundo J, Frade-Sosa B, Sanmartí R. From bench to bedside: Calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front Immunol 2022; 13:1001025. [PMID: 36405711 PMCID: PMC9672845 DOI: 10.3389/fimmu.2022.1001025] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/19/2022] [Indexed: 12/30/2022] Open
Abstract
S100A9/S100A8 (calprotectin), a member of the S100 protein family, has been shown to play a pivotal role in innate immunity activation. Calprotectin plays a critical role in the pathogenesis of rheumatoid arthritis (RA), as it triggers chemotaxis, phagocyte migration and modulation of neutrophils and macrophages. Higher calprotectin levels have been found in synovial fluid, plasma, and serum from RA patients. Recent studies have demonstrated better correlations between serum or plasma calprotectin and composite inflammatory disease activity indexes than c-reactive protein (CRP) or the erythrocyte sedimentation rate (ESR). Calprotectin serum levels decreased after treatment, independently of the DMARD type or strategy. Calprotectin has shown the strongest correlations with other sensitive techniques to detect inflammation, such as ultrasound. Calprotectin independently predicts radiographic progression. However, its value as a biomarker of treatment response and flare after tapering is unclear. This update reviews the current understanding of calprotectin in RA and discusses possible applications as a biomarker in clinical practice.
Collapse
Affiliation(s)
- José Inciarte-Mundo
- Biological aggression and Response Mechanisms, Inflammatory joint diseases (IJDs), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Beatriz Frade-Sosa
- Rheumatology Department, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Raimon Sanmartí
- Biological aggression and Response Mechanisms, Inflammatory joint diseases (IJDs), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain,Rheumatology Department, Hospital Clinic, University of Barcelona, Barcelona, Spain,*Correspondence: Raimon Sanmartí,
| |
Collapse
|
33
|
Hanata N, Ota M, Tsuchida Y, Nagafuchi Y, Okamura T, Shoda H, Fujio K. Serum extracellular traps associate with the activation of myeloid cells in SLE patients with the low level of anti-DNA antibodies. Sci Rep 2022; 12:18397. [PMID: 36319843 PMCID: PMC9626644 DOI: 10.1038/s41598-022-23076-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are involved in systemic lupus erythematosus (SLE). We sought to cluster SLE patients based on serum NET levels. Serum NET levels were higher in SLE patients than healthy controls. Frequencies of pleuritis and myositis were increased in patients with high serum NET levels. Serum NET levels negatively correlated with anti-double stranded DNA (anti-dsDNA) antibody titers and C1q-binding immune complexes, but positively correlated with C-reactive protein (CRP) and monocyte counts. Neutrophil transcriptome analysis demonstrated no difference in NET-associated signatures, irrespective of serum NET levels, suggesting anti-dsDNA antibody-mediated clearance of NETs. In serum, NET levels were significantly correlated with myeloid cell-derived inflammatory molecules. Serum NET-based cluster analysis revealed 3 groups of patients based on serum NET and CRP levels, anti-dsDNA antibody titers, and monocyte count. Monocytes were consistently activated following NET-containing immune complex (NET-IC) stimulation. In conclusion, SLE patients with high serum NET levels had lower anti-dsDNA antibody titers and higher inflammatory responses. NET-IC-stimulated monocytes might associate with an inflammatory response characterized by elevated CRP levels. These findings can apply to precision medicine, as inflammatory processes, rather than antibody-dependent processes, can be targeted in specific subpopulations of SLE patients.
Collapse
Affiliation(s)
- Norio Hanata
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mineto Ota
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- grid.26999.3d0000 0001 2151 536XDepartment of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- grid.26999.3d0000 0001 2151 536XDepartment of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V, Silvestre-Roig C. Neutrophil extracellular traps: from physiology to pathology. Cardiovasc Res 2022; 118:2737-2753. [PMID: 34648022 PMCID: PMC9586562 DOI: 10.1093/cvr/cvab329] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
At the frontline of the host defence response, neutrophil antimicrobial functions have adapted to combat infections and injuries of different origins and magnitude. The release of web-like DNA structures named neutrophil extracellular traps (NETs) constitutes an important mechanism by which neutrophils prevent pathogen dissemination or deal with microorganisms of a bigger size. At the same time, nuclear and granule proteins with microbicidal activity bind to these DNA structures promoting the elimination of entrapped pathogens. However, these toxic properties may produce unwanted effects in the host, when neutrophils uncontrollably release NETs upon persistent inflammation. As a consequence, NET accumulation can produce vessel occlusion, tissue damage, and prolonged inflammation associated with the progression and exacerbation of multiple pathologic conditions. This review outlines recent advances in understanding the mechanisms of NET release and functions in sterile disease. We also discuss mechanisms of physiological regulation and the importance of neutrophil heterogeneity in NET formation and composition.
Collapse
Affiliation(s)
- Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Oliver Soehnlein
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149, Münster, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Solnavägen 1, 171 77, Stockholm, Sweden
| | - Iker Valle Aramburu
- Laboratory of Antimicrobial Defence, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Carlos Silvestre-Roig
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149, Münster, Germany
| |
Collapse
|
35
|
Korolkova AA, Khizha VV, Kozlova DI, Maslyanskiy AL, Vavilova TV. Сalprotectin in the blood plasma as a new biomarker for assessing the activity of rheumatoid arthritis. BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-59-66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aim. To study the potential use and information value of calprotectin in the blood plasma as a new biomarker for determining the activity of rheumatoid arthritis (RA).Materials and methods. The study included 113 people. The treatment group consisted of 79 patients diagnosed with RA; the average age was 58 (± 11.66) years, the median duration of the disease was 10 [6; 15] years. The control group encompassed 34 healthy volunteers; the average age was 40 (± 11.14) years. RA activity was determined according to the Disease Activity Score (DAS) 28 and the Clinical Disease Activity Index (CDAI). The concentration of calprotectin in the blood plasma was determined by the solid-phase enzyme-linked immunosorbent assay. The obtained results were compared with laboratory and clinical parameters, as well as with composite indices (DAS28, CDAI) of RA activity. For mathematical data processing, Spearman’s rank correlation coefficient, linear discriminant analysis, and ROC analysis were used.Results. In the group of patients with RA, the level of calprotectin in the blood was higher than in the control group. A statistically significant relationship was revealed between the level of calprotectin in the blood and all standard parameters of RA activity. The ROC analysis showed that the sensitivity, specificity, and diagnostic accuracy in assessing articular syndrome, as well as moderate and high RA activity according to the composite indices DAS28 and CDAI were higher for calprotectin than for erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). The linear discriminant analysis showed that a combination of ESR and calprotectin levels was the most informative; following it, the probability of correct classification of RA activity, according to the DAS28 index, was 71%. For the CDAI index, only one marker, calprotectin, resulted in a statistically significant classification with a probability of 70.5 %.Conclusion. Сalprotectin in the blood plasma is a promising laboratory biomarker for assessing synovitis activity in RA demonstrating higher accuracy, sensitivity, and specificity than traditional acute-phase reactants.
Collapse
Affiliation(s)
| | - V. V. Khizha
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences
| | - D. I. Kozlova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences
| | | | | |
Collapse
|
36
|
Michailidou D, Duvvuri B, Kuley R, Cuthbertson D, Grayson PC, Khalidi NA, Koening CL, Langford CA, McAlear CA, Moreland LW, Pagnoux C, Seo P, Specks U, Sreih AG, Warrington KJ, Mustelin T, Monach PA, Merkel PA, Lood C. Neutrophil activation in patients with anti-neutrophil cytoplasmic autoantibody-associated vasculitis and large-vessel vasculitis. Arthritis Res Ther 2022; 24:160. [PMID: 35768848 PMCID: PMC9241246 DOI: 10.1186/s13075-022-02849-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/16/2022] [Indexed: 11/23/2022] Open
Abstract
Objective To assess markers of neutrophil activation such as calprotectin and N-formyl methionine (fMET) in anti-neutrophil cytoplasmic autoantibody-associated vasculitis (AAV) and large-vessel vasculitis (LVV). Methods Levels of fMET, and calprotectin, were measured in the plasma of healthy controls (n=30) and patients with AAV (granulomatosis with polyangiitis (GPA, n=123), microscopic polyangiitis (MPA, n=61)), and LVV (Takayasu’s arteritis (TAK, n=58), giant cell arteritis (GCA, n=68)), at times of remission or flare. Disease activity was assessed by physician global assessment. In vitro neutrophil activation assays were performed in the presence or absence of formyl peptide receptor 1 (FPR1) inhibitor cyclosporine H. Results Levels of calprotectin, and fMET were elevated in patients with vasculitis as compared to healthy individuals. Levels of fMET correlated with markers of systemic inflammation: C-reactive protein (r=0.82, p<0.0001), and erythrocyte sedimentation rate (r=0.235, p<0.0001). The neutrophil activation marker, calprotectin was not associated with disease activity. Circulating levels of fMET were associated with neutrophil activation (p<0.01) and were able to induce de novo neutrophil activation via FPR1-mediated signaling. Conclusion Circulating fMET appears to propagate neutrophil activation in AAV and LVV. Inhibition of fMET-mediated FPR1 signaling could be a novel therapeutic intervention for systemic vasculitides.
Collapse
Affiliation(s)
- Despina Michailidou
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Bhargavi Duvvuri
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Runa Kuley
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - David Cuthbertson
- Health Informatics Institute, University of South Florida, South Florida, Tampa, FL, USA
| | - Peter C Grayson
- Systemic Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - Nader A Khalidi
- Division of Rheumatology, Mc Master University, Hamilton, Ontario, Canada
| | - Curry L Koening
- Division of Rheumatology, University of Utah, Salt Lake City, UT, USA
| | | | - Carol A McAlear
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | - Larry W Moreland
- Division of Rheumatology and Clinical Immunology, University of Colorado, Denver, CO, USA
| | | | - Philip Seo
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Antoine G Sreih
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA
| | - Paul A Monach
- Division of Rheumatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter A Merkel
- Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Lood
- Division of Rheumatology, Department of Medicine, University of Washington, 750 Republican Street, Seattle, WA, 98109, USA.
| |
Collapse
|
37
|
Cai ML, Gui L, Huang H, Zhang YK, Zhang L, Chen Z, Sheng YJ. Proteomic Analyses Reveal Higher Levels of Neutrophil Activation in Men Than in Women With Systemic Lupus Erythematosus. Front Immunol 2022; 13:911997. [PMID: 35799787 PMCID: PMC9254905 DOI: 10.3389/fimmu.2022.911997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/26/2022] [Indexed: 11/15/2022] Open
Abstract
Objective Systemic Lupus Erythematosus (SLE) is a systemic autoimmune disease that displays a significant gender difference in terms of incidence and severity. However, the underlying mechanisms accounting for sexual dimorphism remain unclear. The aim of this work was to reveal the heterogeneity in the pathogenesis of SLE between male and female patients. Methods PBMC were collected from 15 patients with SLE (7 males, 8 females) and 15 age-matched healthy controls (7 males, 8 females) for proteomic analysis. The proteins of interest were validated in independent samples (6 male SLE, 6 female SLE). Biomarkers for neutrophil activation (calprotectin), neutrophil extracellular traps (cell-free DNA and elastase), and reactive oxygen species (glutathione) were measured, using enzyme-linked immunosorbent assay, in plasma obtained from 52 individuals. Results Enrichment analysis of proteomic data revealed that type I interferon signaling and neutrophil activation networks mapped to both male and female SLE, while male SLE has a higher level of neutrophil activation compared with female SLE. Western blot validated that PGAM1, BST2, and SERPINB10 involved in neutrophil activation are more abundant in male SLE than in female SLE. Moreover, biomarkers of neutrophil activation and reactive oxygen species were increased in male SLE compared with female SLE. Conclusion Type I interferon activation is a common signature in both male and female SLE, while neutrophil activation is more prominent in male SLE compared with female SLE. Our findings define gender heterogeneity in the pathogenesis of SLE and may facilitate the development of gender-specific treatments.
Collapse
Affiliation(s)
- Ming-long Cai
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lan Gui
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - He Huang
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Yu-kun Zhang
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Li Zhang
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Zhu Chen
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu-jun Sheng
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei, China
- *Correspondence: Yu-jun Sheng,
| |
Collapse
|
38
|
Petchakup C, Yang H, Gong L, He L, Tay HM, Dalan R, Chung AJ, Li KHH, Hou HW. Microfluidic Impedance-Deformability Cytometry for Label-Free Single Neutrophil Mechanophenotyping. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104822. [PMID: 35253966 DOI: 10.1002/smll.202104822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/03/2022] [Indexed: 06/14/2023]
Abstract
The intrinsic biophysical states of neutrophils are associated with immune dysfunctions in diseases. While advanced image-based biophysical flow cytometers can probe cell deformability at high throughput, it is nontrivial to couple different sensing modalities (e.g., electrical) to measure other critical cell attributes including cell viability and membrane integrity. Herein, an "optics-free" impedance-deformability cytometer for multiparametric single cell mechanophenotyping is reported. The microfluidic platform integrates hydrodynamic cell pinching, and multifrequency impedance quantification of cell size, deformability, and membrane impedance (indicative of cell viability and activation). A newly-defined "electrical deformability index" is validated by numerical simulations, and shows strong correlations with the optical cell deformability index of HL-60 experimentally. Human neutrophils treated with various biochemical stimul are further profiled, and distinct differences in multimodal impedance signatures and UMAP analysis are observed. Overall, the integrated cytometer enables label-free cell profiling at throughput of >1000 cells min-1 without any antibodies labeling to facilitate clinical diagnostics.
Collapse
Affiliation(s)
- Chayakorn Petchakup
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Haoning Yang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lingyan Gong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Linwei He
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Hui Min Tay
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Rinkoo Dalan
- Endocrinology Department, Tan Tock Seng Hospital, 11 Jln Tan Tock Seng Road, Singapore, 308433, Singapore
| | - Aram J Chung
- School of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, Republic of Korea
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building Level 11, Singapore, 308232, Singapore
| |
Collapse
|
39
|
Kuley R, Stultz RD, Duvvuri B, Wang T, Fritzler MJ, Hesselstrand R, Nelson JL, Lood C. N-Formyl Methionine Peptide-Mediated Neutrophil Activation in Systemic Sclerosis. Front Immunol 2022; 12:785275. [PMID: 35069556 PMCID: PMC8766990 DOI: 10.3389/fimmu.2021.785275] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Exaggerated neutrophil activation and formation of neutrophil extracellular traps (NETs) are reported in systemic sclerosis (SSc) but its involvement in SSc pathogenesis is not clear. In the present study we assessed markers of neutrophil activation and NET formation in SSc patients in relation to markers of inflammation and disease phenotype. Factors promoting neutrophil activation in SSc remain largely unknown. Among the neutrophil activating factors, mitochondrial-derived N-formyl methionine (fMet) has been reported in several autoinflammatory conditions. The aim of the current study is to assess whether SSc patients have elevated levels of fMet and the role of fMet in neutrophil-mediated inflammation on SSc pathogenesis. Markers of neutrophil activation (calprotectin, NETs) and levels of fMet were analyzed in plasma from two SSc cohorts (n=80 and n=20, respectively) using ELISA. Neutrophil activation assays were performed in presence or absence of formyl peptide receptor 1 (FPR1) inhibitor cyclosporin H. Elevated levels of calprotectin and NETs were observed in SSc patients as compared to healthy controls (p<0.0001) associating with SSc clinical disease characteristics. Further, SSc patients had elevated levels of circulating fMet as compared to healthy controls (p<0.0001). Consistent with a role for fMet-mediated neutrophil activation, fMet levels correlated with levels of calprotectin and NETs (r=0.34, p=0.002; r=0.29, p<0.01 respectively). Additionally, plasma samples from SSc patients with high levels of fMet induced de novo neutrophil activation through FPR1-dependent mechanisms. Our data for the first time implicates an important role for the mitochondrial component fMet in promoting neutrophil-mediated inflammation in SSc.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Ryan D Stultz
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Bhargavi Duvvuri
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Ting Wang
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Marvin J Fritzler
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Roger Hesselstrand
- Department of Clinical Sciences Lund University, Section of Rheumatology, Lund, Sweden
| | - J Lee Nelson
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| |
Collapse
|
40
|
Hematopoiesis, Inflammation and Aging-The Biological Background and Clinical Impact of Anemia and Increased C-Reactive Protein Levels on Elderly Individuals. J Clin Med 2022; 11:jcm11030706. [PMID: 35160156 PMCID: PMC8836692 DOI: 10.3390/jcm11030706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Anemia and systemic signs of inflammation are common in elderly individuals and are associated with decreased survival. The common biological context for these two states is then the hallmarks of aging, i.e., genomic instability, telomere shortening, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication. Such aging-associated alterations of hematopoietic stem cells are probably caused by complex mechanisms and depend on both the aging of hematopoietic (stem) cells and on the supporting stromal cells. The function of inflammatory or immunocompetent cells is also altered by aging. The intracellular signaling initiated by soluble proinflammatory mediators (e.g., IL1, IL6 and TNFα) is altered during aging and contributes to the development of both the inhibition of erythropoiesis with anemia as well as to the development of the acute-phase reaction as a systemic sign of inflammation with increased CRP levels. Both anemia and increased CRP levels are associated with decreased overall survival and increased cardiovascular mortality. The handling of elderly patients with inflammation and/or anemia should in our opinion be individualized; all of them should have a limited evaluation with regard to the cause of the abnormalities, but the extent of additional and especially invasive diagnostic evaluation should be based on an overall clinical evaluation and the possible therapeutic consequences.
Collapse
|
41
|
Zhao J, Jiang P, Guo S, Schrodi SJ, He D. Apoptosis, Autophagy, NETosis, Necroptosis, and Pyroptosis Mediated Programmed Cell Death as Targets for Innovative Therapy in Rheumatoid Arthritis. Front Immunol 2022; 12:809806. [PMID: 35003139 PMCID: PMC8739882 DOI: 10.3389/fimmu.2021.809806] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/13/2021] [Indexed: 01/13/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease that can lead to clinical manifestations of systemic diseases. Its leading features include chronic synovial inflammation and degeneration of the bones and joints. In the past decades, multiple susceptibilities for rheumatoid arthritis have been identified along with the development of a remarkable variety of drugs for its treatment; which include analgesics, glucocorticoids, nonsteroidal anti-inflammatory medications (NSAIDs), disease-modifying anti-rheumatic drugs (DMARDs), and biologic response modifiers (bDMARDs). Despite the existence of many clinical treatment options, the prognosis of some patients remains poor due to complex mechanism of the disease. Programmed cell death (PCD) has been extensively studied and ascertained to be one of the essential pathological mechanisms of RA. Its dysregulation in various associated cell types contributes to the development of RA. In this review, we summarize the role of apoptosis, cell death-associated neutrophil extracellular trap formation, necroptosis, pyroptosis, and autophagy in the pathophysiology of RA to provide a theoretical reference and insightful direction to the discovery and development of novel therapeutic targets for RA.
Collapse
Affiliation(s)
- Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Steven J Schrodi
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
42
|
Polymorphonuclear Neutrophils in Rheumatoid Arthritis and Systemic Lupus Erythematosus: More Complicated Than Anticipated. IMMUNO 2022. [DOI: 10.3390/immuno2010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Polymorphonuclear neutrophils (PMN) are the most abundant leucocytes in the circulation in humans. They represent a heterogeneous population exerting diverse functions through several activities. Usually described as typical pro-inflammatory cells, immunomodulatory properties of PMNs have been reported. Among others, once activated and depending on the stimulus, PMNs expel neutrophil extracellular traps (NET) in the extracellular space. NETs are complexes made of DNA and granule proteins representing an innate immune mechanism fighting infections. Nevertheless, an excess of NET formation might be involved in the development of inflammatory or autoimmune responses. Systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are two chronic, inflammatory, autoimmune diseases of unknown etiology and affecting mostly women. Several abnormal or non-classical functions of PMNs or PMN sub-populations have been described in SLE and RA. Particularly, NETs have been suggested to trigger pro-inflammatory responses by exposing pro-inflammatory mediators. Likewise, NETs may be the targets of autoantibodies or even might trigger the development of autoantibodies by exposing autoantigens. In the present review, we will summarize heterogeneous properties of human PMNs and we will discuss recent evidence linking PMNs and NETs to the pathogenesis of both SLE and RA.
Collapse
|
43
|
Wang X, Hefton A, Ni K, Ukadike KC, Bowen MA, Eckert M, Stevens A, Lood C, Mustelin T. Autoantibodies Against Unmodified and Citrullinated Human Endogenous Retrovirus K Envelope Protein in Patients With Rheumatoid Arthritis. J Rheumatol Suppl 2022; 49:26-35. [PMID: 34334364 PMCID: PMC8963793 DOI: 10.3899/jrheum.201492] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Autoantibodies against proteins encoded by human endogenous retrovirus K (HERV-K) have been reported in patients with rheumatoid arthritis (RA), but their relevance, if any, has remained unresolved. We revisited this question and tested if such autoantibodies may react with citrullinated epitopes on the envelope (Env) protein of HERV-K. METHODS Immunoblotting and ELISAs were conducted with unmodified Env protein and with Env citrullinated by protein arginine deiminase 4 (PAD4). Sera from 100 patients with RA, plasma from 32 patients with juvenile idiopathic arthritis (JIA), and healthy adult and pediatric controls were included. Antibody reactivity was evaluated for correlations with clinical and laboratory variables of the patients. RESULTS We replicated and expanded upon published data suggesting that patients with RA or JIA have autoantibodies against HERV-K Env, some with high titers. Anti-HERV-K antibodies correlated with cigarette smoking and with circulating myeloperoxidase-DNA complexes indicative of nonapoptotic neutrophil cell death. Further, most of the patients with RA, but not those with JIA, had autoantibodies that reacted more strongly with Env that was citrullinated by PAD4. These anticitrullinated Env autoantibodies correlated with seropositivity and tended to be higher in patients with erosive disease. CONCLUSION Our data suggest that anti-HERV-K immunity is elevated in RA and JIA and may have a connection with pathogenic protein citrullination in RA.
Collapse
Affiliation(s)
- Xiaoxing Wang
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| | - Amanda Hefton
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| | - Kathryn Ni
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| | - Kennedy C. Ukadike
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| | - Michael A. Bowen
- M.A. Bowen, PhD, Product and Process Development, Allogene Therapeutics, San Francisco, California
| | - Mary Eckert
- M. Eckert, BS, Seattle Children’s Research Institute, Seattle, Washington
| | - Anne Stevens
- A. Stevens, MD, Professor, Seattle Children’s Research Institute, Seattle, Division of Rheumatology, Department of Pediatrics, University of Washington, Seattle, Washington, and Janssen Research & Development, LLC, Wayne, Pennsylvania, USA
| | - Christian Lood
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| | - Tomas Mustelin
- X. Wang, PhD, A. Hefton, K. Ni, BS, K.C. Ukadike, MD, Acting Instructor, C. Lood, PhD, Associate Professor, T. Mustelin, MD, PhD, Professor, Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
44
|
Guo Y, Gao F, Wang X, Pan Z, Wang Q, Xu S, Pan S, Li L, Zhao D, Qian J. Spontaneous formation of neutrophil extracellular traps is associated with autophagy. Sci Rep 2021; 11:24005. [PMID: 34907287 PMCID: PMC8671464 DOI: 10.1038/s41598-021-03520-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 12/17/2022] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs), via NETosis, as a defense mechanism against pathogens. Neutrophils can release NETs spontaneously; however, the mechanisms underlying spontaneous NETosis remain unclear. Neutrophils isolated from healthy donors were tested for NET formation and autophagy at 1, 6, 12, and 24 h after incubation. Autophagy response was evaluated in response to various autophagy inducers and inhibitors. The relationship between autophagy and NETosis was detected in vivo using an ovalbumin-induced mouse model of asthma. We found that the increase in the proportion of spontaneous NETosis was time-dependent. The number of autophagy-positive cells also increased over time and LC3B protein played an integral role in NET formation. Trehalose (an inducer of mTOR-independent autophagy) treatment significantly increased NET formation, whereas rapamycin (an mTOR-dependent autophagy inducer) did not increase NET release by neutrophils. Compared with the control group, 3-methyladenine (an autophagy sequestration inhibitor) and hydroxychloroquine sulfate (autophagosome-lysosome fusion inhibitor) treatments significantly reduced the percentage of NET-positive cells. In vivo studies on ovalbumin-induced asthma lung sections revealed NETs and LC3B and citH3 proteins were found to co-localize with DNA. Our findings suggest that autophagy plays a crucial role in aging-related spontaneous NETosis.
Collapse
Affiliation(s)
- Yun Guo
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Fei Gao
- Department of Intensive Care Unit, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xin Wang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Zhenzhen Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Qian Wang
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shiyao Xu
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shanshan Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Ling Li
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China.
| | - Jun Qian
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
45
|
O'Neil LJ, Hu P, Liu Q, Islam MM, Spicer V, Rech J, Hueber A, Anaparti V, Smolik I, El-Gabalawy HS, Schett G, Wilkins JA. Proteomic Approaches to Defining Remission and the Risk of Relapse in Rheumatoid Arthritis. Front Immunol 2021; 12:729681. [PMID: 34867950 PMCID: PMC8636686 DOI: 10.3389/fimmu.2021.729681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022] Open
Abstract
Objectives Patients with Rheumatoid Arthritis (RA) are increasingly achieving stable disease remission, yet the mechanisms that govern ongoing clinical disease and subsequent risk of future flare are not well understood. We sought to identify serum proteomic alterations that dictate clinically important features of stable RA, and couple broad-based proteomics with machine learning to predict future flare. Methods We studied baseline serum samples from a cohort of stable RA patients (RETRO, n = 130) in clinical remission (DAS28<2.6) and quantified 1307 serum proteins using the SOMAscan platform. Unsupervised hierarchical clustering and supervised classification were applied to identify proteomic-driven clusters and model biomarkers that were associated with future disease flare after 12 months of follow-up and RA medication withdrawal. Network analysis was used to define pathways that were enriched in proteomic datasets. Results We defined 4 proteomic clusters, with one cluster (Cluster 4) displaying a lower mean DAS28 score (p = 0.03), with DAS28 associating with humoral immune responses and complement activation. Clustering did not clearly predict future risk of flare, however an XGboost machine learning algorithm classified patients who relapsed with an AUC (area under the receiver operating characteristic curve) of 0.80 using only baseline serum proteomics. Conclusions The serum proteome provides a rich dataset to understand stable RA and its clinical heterogeneity. Combining proteomics and machine learning may enable prediction of future RA disease flare in patients with RA who aim to withdrawal therapy.
Collapse
Affiliation(s)
- Liam J O'Neil
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Qian Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Md Mohaiminul Islam
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Juergen Rech
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Axel Hueber
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Vidyanand Anaparti
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Irene Smolik
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Hani S El-Gabalawy
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Georg Schett
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - John A Wilkins
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| |
Collapse
|
46
|
Fu X, Liu H, Huang G, Dai SS. The emerging role of neutrophils in autoimmune-associated disorders: effector, predictor, and therapeutic targets. MedComm (Beijing) 2021; 2:402-413. [PMID: 34766153 PMCID: PMC8554667 DOI: 10.1002/mco2.69] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/19/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are essential components of the immune system and have vital roles in the pathogenesis of autoimmune disorders. As effector cells, neutrophils promote autoimmune disease by releasing cytokines and chemokines cascades that accompany inflammation, neutrophil extracellular traps (NETs) regulating immune responses through cell-cell interactions. More recent evidence has extended functions of neutrophils. Accumulating evidence implicated neutrophils contribute to tissue damage during a broad range of disorders, involving rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), primary sjögren's syndrome (pSS), multiple sclerosis (MS), crohn's disease (CD), and gout. A variety of studies have reported on the functional role of neutrophils as therapeutic targets in autoimmune diseases. However, challenges and controversies in the field remain. Enhancing our understanding of neutrophils' role in autoimmune disorders may further advance the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Xiaohong Fu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science Third Military Medical University (Army Medical University) Chongqing China
| | - Heting Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science Third Military Medical University (Army Medical University) Chongqing China
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science Third Military Medical University (Army Medical University) Chongqing China
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science Third Military Medical University (Army Medical University) Chongqing China
| |
Collapse
|
47
|
Mun Y, Hwang JS, Shin YJ. Role of Neutrophils on the Ocular Surface. Int J Mol Sci 2021; 22:10386. [PMID: 34638724 PMCID: PMC8508808 DOI: 10.3390/ijms221910386] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
The ocular surface is a gateway that contacts the outside and receives stimulation from the outside. The corneal innate immune system is composed of many types of cells, including epithelial cells, fibroblasts, natural killer cells, macrophages, neutrophils, dendritic cells, mast cells, basophils, eosinophils, mucin, and lysozyme. Neutrophil infiltration and degranulation occur on the ocular surface. Degranulation, neutrophil extracellular traps formation, called NETosis, and autophagy in neutrophils are involved in the pathogenesis of ocular surface diseases. It is necessary to understand the role of neutrophils on the ocular surface. Furthermore, there is a need for research on therapeutic agents targeting neutrophils and neutrophil extracellular trap formation for ocular surface diseases.
Collapse
Affiliation(s)
- Yongseok Mun
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| |
Collapse
|
48
|
La C, Lê PQ, Ferster A, Goffin L, Spruyt D, Lauwerys B, Durez P, Boulanger C, Sokolova T, Rasschaert J, Badot V. Serum calprotectin (S100A8/A9): a promising biomarker in diagnosis and follow-up in different subgroups of juvenile idiopathic arthritis. RMD Open 2021; 7:rmdopen-2021-001646. [PMID: 34108235 PMCID: PMC8191626 DOI: 10.1136/rmdopen-2021-001646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022] Open
Abstract
Introduction In the management of juvenile idiopathic arthritis (JIA), there is a lack of diagnostic and prognostic biomarkers. This study assesses the use of serum calprotectin (sCal) as a marker to monitor disease activity, and as a classification and prognosis tool of response to treatment or risk of flares in patients with JIA. Methods Eighty-one patients with JIA from the CAP48 multicentric cohort were included in this study, as well as 11 non-paediatric healthy controls. An ELISA method was used to quantify sCal with a commercial kit. Results Patients with an active disease compared with healthy controls and with patients with inactive disease showed an eightfold and a twofold increased level of sCal, respectively. sCal was found to be correlated with the C-reactive protein (CRP) and even more strongly with the erythrocyte sedimentation rate. Evolution of DAS28 scores correlated well with evolution of sCal, as opposed to evolution of CRP. With regard to CRP, sCal could differentiate forms with active oligoarthritis from polyarthritis and systemic forms. However, sCal brought an added value compared with the CRP as a prognosis marker. Indeed, patients with active disease and reaching minimal disease activity (according to Juvenile Arthritis Disease Activity Score) at 6 months following the test had higher sCal levels, while patients with inactive disease had higher sCal levels if a flare was observed up to 3–9 months following the test. Conclusions This study confirms the potential uses of sCal as a biomarker in the diagnosis and follow-up of JIA.
Collapse
Affiliation(s)
- Céline La
- Department of Rheumatology, CHU Brugmann, Bruxelles, Belgium .,Department of Pediatric Rheumatology, Hôpital Universitaire des Enfants Reine Fabiola, Bruxelles, Belgium.,Department of Rheumatology, Hôpital Erasme, Bruxelles, Belgium
| | - Phu Quoc Lê
- Department of Pediatric Rheumatology, Hôpital Universitaire des Enfants Reine Fabiola, Bruxelles, Belgium
| | - Alina Ferster
- Department of Pediatric Rheumatology, Hôpital Universitaire des Enfants Reine Fabiola, Bruxelles, Belgium
| | - Laurence Goffin
- Department of Pediatric Rheumatology, Hôpital Universitaire des Enfants Reine Fabiola, Bruxelles, Belgium
| | - Delphine Spruyt
- Laboratory of Bone and Metabolic Biochemistry, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Bernard Lauwerys
- Department of Rheumatology, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Patrick Durez
- Department of Rheumatology, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Cecile Boulanger
- Department of Rheumatology, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Tatiana Sokolova
- Institut de Recherche expérimentale et Clinique (IREC), Université catholique de Louvain Secteur des sciences de la santé, Bruxelles, Belgium
| | - Joanne Rasschaert
- Laboratory of Bone and Metabolic Biochemistry, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Valérie Badot
- Department of Rheumatology, CHU Brugmann, Bruxelles, Belgium
| |
Collapse
|
49
|
Vedder D, Gerritsen M, Nurmohamed MT, van Vollenhoven RF, Lood C. A neutrophil signature is strongly associated with increased cardiovascular risk in gout. Rheumatology (Oxford) 2021; 60:2783-2790. [PMID: 33188698 PMCID: PMC8213432 DOI: 10.1093/rheumatology/keaa712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To investigate the association between neutrophil activation and cardiovascular risk in gout patients. We hypothesize that neutrophil activation mediates inflammation and therefore takes part in atherogenesis in gout patients. METHOD Patient data were collected from 75 consecutive gout patients participating in the Reade gout cohort Amsterdam. Levels of neutrophil extracellular traps (NETs) and neutrophil activation (calprotectin and peroxidase activity) were analysed by ELISA and fluorimetry in plasma and compared with healthy controls. Markers of neutrophil activation were related to clinical markers of cardiovascular risk, including BMI, smoking, blood pressure, lipid profile and 10 year risk of cardiovascular mortality (EU-SCORE). RESULTS Increased levels of NETs were found in gout patients, although increased levels were not associated with cardiovascular risk. However, markers of neutrophil activation, including peroxidase activity correlated with waist:hip ratio (β = 0.33, P < 0.001), cholesterol ratio (β = 0.46, P < 0.005) and triglycerides (β = 0.60, P < 0.001) as well as the 10 year risk of cardiovascular mortality (β = 0.44, P = 0.001). Calprotectin levels were elevated in hypertension (P = 0.005) and diabetes (P = 0.02). Finally, gout patients with high levels of both peroxidase and calprotectin ('neutrophil activation signature') had a markedly elevated cardiovascular risk score (P = 0.001), with 68% of the patients having high cardiovascular risk (odds ratio 2.9, P = 0.03). CONCLUSION We demonstrated elevated levels of neutrophil activation markers, MPO and calprotectin in gout patients as compared with healthy controls. Of note, neutrophil activation markers were associated with several risk factors for cardiovascular disease, including hyperlipidaemia, hypertension and diabetes. Finally, the presence of a neutrophil activation signature was strongly associated with an increased 10 year risk of cardiovascular mortality. Further studies are needed to determine whether gout-specific factors and/or cardiovascular risk factors contribute to the elevated neutrophil activation observed in these patients.
Collapse
Affiliation(s)
- Daisy Vedder
- Amsterdam Rheumatology and Immunology Center Reade, Amsterdam, The Netherlands.,Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Martijn Gerritsen
- Amsterdam Rheumatology and Immunology Center Reade, Amsterdam, The Netherlands
| | - Michael T Nurmohamed
- Amsterdam Rheumatology and Immunology Center Reade, Amsterdam, The Netherlands.,Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Ronald F van Vollenhoven
- Amsterdam Rheumatology and Immunology Center Reade, Amsterdam, The Netherlands.,Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
50
|
Abstract
Based on the PubMed data, we have been performing a yearly evaluation of the publications related to autoimmune diseases and immunology to ascertain the relative weight of the former in the scientific literature. It is particularly intriguing to observe that despite the numerous new avenues of immune-related mechanisms, such as cancer immunotherapy, the proportion of immunology manuscripts related to autoimmunity continues to increase and has been approaching 20% in 2019. As in the previous 13 years, we performed an arbitrary selection of the peer-reviewed articles published by the major dedicated Journals and discussed the common themes which continue to outnumber peculiarites in autoimmune diseases. The investigated areas included systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriatic arthritis (PsA), autoantibodies (autoAbs), and common therapeutic avenues and novel pathogenic mechanisms for autoimmune conditions. Some examples include new pathogenetic evidence which is well represented by IL21 or P2X7 receptor (P2X7R) in SLE or the application of single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq), and flow cytometry for the analysis of different cellular populations in RA. Cumulatively and of interest to the clinicians, a large number of findings continue to underline the importance of a strict relationship between basic and clinical science to define new pathogenetic and therapeutic developments. The therapeutic pipeline in autoimmunity continues to grow and maintain a constant flow of new molecules, as well illustrated in RA and PsA, and this is most certainly derived from the new basic evidence and the high-throughput tools applied to autoimmune diseases.
Collapse
|