1
|
Zeng Y, Xu W, Chao P, Xiao Y, Yang T. Neutrophil extracellular traps as a potential marker of systemic lupus erythematosus activity. Int Immunopharmacol 2025; 146:113840. [PMID: 39689598 DOI: 10.1016/j.intimp.2024.113840] [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: 07/27/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND The heterogeneity of systemic lupus erythematosus (SLE) poses a significant challenge in identifying biomarkers for assessing disease activity. Currently, there is a paucity of established biomarkers capable of evaluating SLE flares. This study aimed to identify novel biomarkers that exhibit improved diagnostic accuracy in assessing SLE activity. METHODS A cross-sectional study was conducted at Zhongshan Hospital Xiamen University from August 2021 to April 2024,enrolling 118 patients with SLE, including 81 cases of active SLE, 50 cases of active lupus nephritis (LN) and 30 cases of active non-LN. The objective was to evaluate the diagnostic accuracy of novel biomarker called Neutrophil Extracellular Traps(NETs) for SLE activity and analyze its correlations with conventional biomarkers such as complement C3, C4, and anti-dsDNA. RESULTS Serum NETs levels were significantly elevated in patients with active SLE and active LN(P < 0.001). Furthermore, positive correlations were observed between NETs levels and disease activity score based on Systemic Lupus Erythematosus Disease Activity Index-2 K (SLEDAI-2 K) (r = 0.64, P < 0.001), as well as anti-dsDNA antibody (r = 0.54, P < 0.001).Conversely, the NETs levels were negativity correlated with complement C3 concentration (r = -0.50, P < 0.001), as well as C4 concentration (r = -0.34,P < 0.001). Univariate and multivariate analysis revealed two biomarkers performed statistical significance: NETs (OR = 6.802, 95 %CI: 2.414-19.167,P < 0.001) and anti-dsDN A(OR = 3.95,95 %CI:1.582-9.864, P = 0.003). NETs had the highest AUC of 0.82(P < 0.001), with a cut-off at 515.47 ng/L demonstrating 61.63 % sensitivity and 96.87 % specificity. For the active LN group, the AUC was found to be 0.97 (P < 0.001), with a cutoff value of 515.47 ng/L, sensitivity of 100 %, and specificity of 59.76 %. Moreover, the active non-LN group had AUC of 0.70 (P = 0.007), with the same cutoff value, sensitivity of 89.61 %, and specificity of 51.61 %. CONCLUSION In contrast to conventional laboratory markers, serum NETs represent a novel diagnostic marker for assessing disease activity in SLE, demonstrating promising potential for clinical application.
Collapse
Affiliation(s)
- Yanli Zeng
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease,School of Medicine,XiamenUniversity, Xiamen 361004, China; Xiamen Clinical Laboratory Quality ControlCenter,Zhongshan Hospital Xiamen University, Xiamen 361004, China.
| | - Wenlong Xu
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Pengli Chao
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Yun Xiao
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Tianci Yang
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease,School of Medicine,XiamenUniversity, Xiamen 361004, China; Xiamen Clinical Laboratory Quality ControlCenter,Zhongshan Hospital Xiamen University, Xiamen 361004, China.
| |
Collapse
|
2
|
Azzouz D, Palaniyar N. How Do ROS Induce NETosis? Oxidative DNA Damage, DNA Repair, and Chromatin Decondensation. Biomolecules 2024; 14:1307. [PMID: 39456240 PMCID: PMC11505619 DOI: 10.3390/biom14101307] [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: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are intricate, DNA-based, web-like structures adorned with cytotoxic proteins. They play a crucial role in antimicrobial defense but are also implicated in autoimmune diseases and tissue injury. The process of NET formation, known as NETosis, is a regulated cell death mechanism that involves the release of these structures and is unique to neutrophils. NETosis is heavily dependent on the production of reactive oxygen species (ROS), which can be generated either through NADPH oxidase (NOX) or mitochondrial pathways, leading to NOX-dependent or NOX-independent NETosis, respectively. Recent research has revealed an intricate interplay between ROS production, DNA repair, and NET formation in different contexts. UV radiation can trigger a combined process of NETosis and apoptosis, known as apoNETosis, driven by mitochondrial ROS and DNA repair. Similarly, in calcium ionophore-induced NETosis, both ROS and DNA repair are key components, but only play a partial role. In the case of bacterial infections, the early stages of DNA repair are pivotal. Interestingly, in serum-free conditions, spontaneous NETosis occurs through NOX-derived ROS, with early-stage DNA repair inhibition halting the process, while late-stage inhibition increases it. The intricate balance between DNA repair processes and ROS production appears to be a critical factor in regulating NET formation, with different pathways being activated depending on the nature of the stimulus. These findings not only deepen our understanding of the mechanisms behind NETosis but also suggest potential therapeutic targets for conditions where NETs contribute to disease pathology.
Collapse
Affiliation(s)
- Dhia Azzouz
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
3
|
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
|
4
|
Pernaute OS. Voclosporin is on the table, let's taste it. Rheumatology (Oxford) 2024; 63:1762-1765. [PMID: 38224548 DOI: 10.1093/rheumatology/keae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/01/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024] Open
Affiliation(s)
- Olga Sánchez Pernaute
- Unit for Autoimmune Diseases, Rheumatology Department, Fundación Jiménez Díaz University Hospital and Health Research Institute (IIS-FJD), Madrid, Spain
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Lv X, Shang Y, Ning Y, Yu W, Wang J. Pharmacological targets of SGLT2 inhibitors on IgA nephropathy and membranous nephropathy: a mendelian randomization study. Front Pharmacol 2024; 15:1399881. [PMID: 38846092 PMCID: PMC11155304 DOI: 10.3389/fphar.2024.1399881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Emerging research suggests that sodium-glucose cotransporter 2 (SGLT2) inhibitors may play a pivotal role in the treatment of primary glomerular diseases. This study was aimed to investigate potential pharmacological targets connecting SGLT2 inhibitors with IgA nephropathy (IgAN) and membranous nephropathy (MN). Methods A univariate Mendelian randomization (MR) analysis was conducted using publicly available genome-wide association studies (GWAS) datasets. Co-localization analysis was used to identify potential connections between target genes and IgAN and MN. Then, Comparative Toxicogenomics Database (CTD) was employed to predict diseases associated with these target genes and SGLT2 inhibitors (canagliflozin, dapagliflozin, and empagliflozin). Subsequently, phenotypic scan analyses were applied to explore the causal relationships between the predicted diseases and target genes. Finally, we analyzed the immune signaling pathways involving pharmacological target genes using the Kyoto encyclopedia of genes and genomes (KEGG). Results The results of MR analysis revealed that eight drug targets were causally linked to the occurrence of IgAN, while 14 drug targets were linked to MN. In the case of IgAN, LCN2 and AGER emerged as co-localized genes related to the pharmacological agent of dapagliflozin and the occurrence of IgAN. LCN2 was identified as a risk factor, while AGER was exhibited a protective role. KEGG analysis revealed that LCN2 is involved in the interleukin (IL)-17 immune signaling pathway, while AGER is associated with the neutrophil extracellular traps (NETs) signaling immune pathway. No positive co-localization results of the target genes were observed between two other SGLT2 inhibitors (canagliflozin and empagliflozin) and the occurrence of IgAN, nor between the three SGLT2 inhibitors and the occurrence of MN. Conclusion Our study provided evidence supporting a causal relationship between specific SGLT2 inhibitors and IgAN. Furthermore, we found that dapagliflozin may act on IgAN through the genes LCN2 and AGER.
Collapse
Affiliation(s)
- Xin Lv
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Shang
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Ning
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weimin Yu
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
8
|
Zhou Y, Deng S, Du C, Zhang L, Li L, Liu Y, Wang Y, Zhang Y, Zhu L. Leukotriene B4-induced neutrophil extracellular traps impede the clearance of Pneumocystis. Eur J Immunol 2024; 54:e2350779. [PMID: 38440842 DOI: 10.1002/eji.202350779] [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: 09/20/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Pneumocystis pneumonia (PCP) is a fungal pulmonary disease with high mortality in immunocompromised patients. Neutrophils are essential in defending against fungal infections; however, their role in PCP is controversial. Here we aim to investigate the effects of neutrophil extracellular traps (NETs) on Pneumocystis clearance and lung injury using a mouse model of PCP. Intriguingly, although neutrophils play a fundamental role in defending against fungal infections, NETs failed to eliminate Pneumocystis, but instead impaired the killing of Pneumocystis. Mechanically, Pneumocystis triggered Leukotriene B4 (LTB4)-dependent neutrophil swarming, leading to agglutinative NET formation. Blocking Leukotriene B4 with its receptor antagonist Etalocib significantly reduced the accumulation and NET release of neutrophils in vitro and in vivo, enhanced the killing ability of neutrophils against Pneumocystis, and alleviated lung injury in PCP mice. This study identifies the deleterious role of agglutinative NETs in Pneumocystis infection and reveals a new way to prevent NET formation, which provides new insights into the pathogenesis of PCP.
Collapse
Affiliation(s)
- Yanxi Zhou
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shuwei Deng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chunjing Du
- Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liang Zhang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Lan Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yujia Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yijie Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yue Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liuluan Zhu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Li X, Hu L, Naeem A, Xiao S, Yang M, Shang H, Zhang J. Neutrophil Extracellular Traps in Tumors and Potential Use of Traditional Herbal Medicine Formulations for Its Regulation. Int J Nanomedicine 2024; 19:2851-2877. [PMID: 38529365 PMCID: PMC10961241 DOI: 10.2147/ijn.s449181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular fibers composed of deoxyribonucleic acid (DNA) and decorated proteins produced by neutrophils. Recently, NETs have been associated with the development of many diseases, including tumors. Herein, we reviewed the correlation between NETs and tumors. In addition, we detailed active compounds from traditional herbal medicine formulations that inhibit NETs, related nanodrug delivery systems, and antibodies that serve as "guiding moieties" to ensure targeted delivery to NETs. Furthermore, we discussed the strategies used by pathogenic microorganisms to evade NETs.
Collapse
Affiliation(s)
- Xiang Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Lei Hu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Abid Naeem
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
| | - Shanghua Xiao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Hongming Shang
- Department of Biochemistry & Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Jing Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| |
Collapse
|
10
|
Li Z, Yuan T. Neutrophil extracellular traps in adult diseases and neonatal bacterial infectious diseases: A review. Heliyon 2024; 10:e23559. [PMID: 38173520 PMCID: PMC10761809 DOI: 10.1016/j.heliyon.2023.e23559] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Neutrophils, the most abundant type of white blood cells, are pivotal in fighting bacterial infections due to their immunological and anti-infection capabilities. In recent years, scientists have discovered a novel mechanism known as neutrophil extracellular traps, which are fibrous networks primarily released by neutrophils that combat bacterial infections. There is a growing interest in studying NETs and their role in human infectious diseases, particularly in neonates susceptible to bacterial infections. NETs and their components have been found in various samples from neonatal-infected patients, providing a new route for early diagnosis of neonatal infectious diseases. This paper aims to summarize the studies on NETs in adult diseases and mainly discuss NETs in neonatal sepsis, necrotizing enterocolitis, and purulent meningitis, to provide scientific evidence for early monitoring, diagnosis, and treatment of neonatal infections.
Collapse
Affiliation(s)
- Ziheng Li
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| |
Collapse
|
11
|
Liu B, Deng Y, Duan Z, Chu C, Wang X, Yang C, Li J, Ding W. Neutrophil extracellular traps promote intestinal barrier dysfunction by regulating macrophage polarization during trauma/hemorrhagic shock via the TGF-β signaling pathway. Cell Signal 2024; 113:110941. [PMID: 37890686 DOI: 10.1016/j.cellsig.2023.110941] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/01/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The mechanism by which neutrophil extracellular traps (NETs) may cause intestinal barrier dysfunction in response to trauma/hemorrhagic shock (T/HS) remains unclear. In this study, the roles and mechanisms of NETs in macrophage polarization were examined to determine whether this process plays a role in tissue damage associated with T/HS. Rat models of T/HS and macrophage polarization were developed and the levels of NETs formation in the intestinal tissue of T/HS rats were assessed. NET formation was inhibited in models of T/HS to examine the effect on intestinal inflammation and barrier injury. The proportions of pro-inflammatory and anti-inflammatory macrophages in the damaged intestinal tissues were measured. Finally, high-throughput sequencing was performed to investigate the underlying mechanisms involved in this process. The study revealed that the level of NETs formation was increased and that inhibition of NETs formation alleviated the intestinal inflammation and barrier injury. Moreover, the number of pro-inflammatory macrophages increased and the number of anti-inflammatory macrophages decreased. RNA sequencing analysis indicated that NETs formation decreased the expression of transforming growth factor-beta receptor 2 (TGFBR2), bioinformatic analyses revealed that TGFBR2 was significantly enriched in the transforming growth factor-beta (TGF-β) signaling pathway. Verification experiments showed that NETs impeded macrophage differentiation into the anti-inflammatory/M2 phenotype and inhibited TGFBR2 and TGF-β expression in macrophages. However, treatment with DNase I and overexpression of TGFBR2, and inhibition of TGF-β promoted and prevented this process, respectively. NETs may regulate the macrophage polarization process by promoting intestinal barrier dysfunction in T/HS rats through the TGFBR2-mediated TGF-β signaling pathway.
Collapse
Affiliation(s)
- Baochen Liu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunxuan Deng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zehua Duan
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chengnan Chu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xingyu Wang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao Yang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Weiwei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Xu Y, Lan P, Wang T. The Role of Immune Cells in the Pathogenesis of Idiopathic Pulmonary Fibrosis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1984. [PMID: 38004032 PMCID: PMC10672798 DOI: 10.3390/medicina59111984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown etiology with limited treatment options. The role of the immune system in IPF has received increasing attention. Uncontrolled immune responses drive the onset and progression of IPF. This article provides an overview of the role of innate immune cells (including macrophages, neutrophils, mast cells, eosinophils, dendritic cells, nature killer cells, nature kill cells and γδ T cells) and adaptive immune cells (including Th1 cells, Th2 cells, Th9 cells, Th17 cells, Th22 cells, cytotoxic T cells, B lymphocytes and Treg cells) in IPF. In addition, we review the current status of pharmacological treatments for IPF and new developments in immunotherapy. A deeper comprehension of the immune system's function in IPF may contribute to the development of targeted immunomodulatory therapies that can alter the course of the disease.
Collapse
Affiliation(s)
- Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
13
|
Chen XQ, Tu L, Tang Q, Zou JS, Yun X, Qin YH. DNase I targeted degradation of neutrophil extracellular traps to reduce the damage on IgAV rat. PLoS One 2023; 18:e0291592. [PMID: 37906560 PMCID: PMC10617705 DOI: 10.1371/journal.pone.0291592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/30/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND In the past two years, studies have found a significant increase in neutrophil extracellular traps (NETs) in patients with IgA vasculitis (IgAV), which is correlated with the severity of the disease. NETs have been reported as an intervention target in inflammatory and autoimmune diseases. This study aimed to investigate the effect of targeted degradation of NETs using DNase I in IgAV rat model. METHODS Twenty-four Sprague-Dawley rats were randomly divided into three groups: the IgAV model group, the DNase I intervention group and the normal control group, with an average of 8 rats in each group. The model group was established by using Indian ink, ovalbumin, and Freund's complete adjuvant. In the intervention group, DNase I was injected through tail vein 3 days before the end of established model. The circulating cell free-DNA (cf-DNA) and myeloperoxidase-DNA (MPO-DNA) were analyzed. The presence of NETs in the kidney, gastric antrum and descending duodenum were detected using multiple fluorescences immunohistochemistry and Western blots. Morphological changes of the tissues were observed. RESULTS After the intervention of DNase I, there was a significant reduction in cf-DNA and MPO-DNA levels in the intervention group compared to the IgAV model group (all P<0.001). The presence of NETs in renal, gastric, and duodenal tissues of the intervention group exhibited a significant decrease compared to the IgAV model group (P < 0.01). Moreover, the intervention group demonstrated significantly lower levels of renal MPO and citrullinated histone H3 (citH3) protein expression when compared to the IgAV model group (all P < 0.05). The HE staining results of intervention group demonstrated a significant reduction in congestion within glomerular and interstitial capillaries. Moreover, there was a notable improvement in gastric and intestinal mucosa necrosis, congestion and bleeding. Additionally, there was a substantial decrease in inflammatory cells infiltration. CONCLUSION The degradation of NETs can be targeted by DNase I to mitigate tissue damage in IgAV rat models. Targeted regulation of NETs holds potential as a therapeutic approach for IgAV.
Collapse
Affiliation(s)
- Xiu-Qi Chen
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Li Tu
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Qing Tang
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Jia-Sen Zou
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xiang Yun
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yuan-Han Qin
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
14
|
Goldberg I, Granot G, Telerman A, Partouche S, Shochat T, Halperin E, Gafter-Gvili A, Shargian L, Yeshurun M, Raanani P, Wolach O, Yahalom V. Extracorporeal photopheresis induces NETosis in neutrophils derived from patients with chronic graft-vs-host disease. J Clin Apher 2023; 38:615-621. [PMID: 37439388 DOI: 10.1002/jca.22073] [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: 11/15/2022] [Revised: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Extracorporeal photopheresis (ECP) is considered an effective treatment for patients with chronic graft vs host disease (cGVHD) and demonstrates efficacy in ameliorating GVHD. The mechanism by which ECP acts against cGVHD is not fully understood. Preliminary observations have hinted at the potential involvement of neutrophil extracellular traps (NETs) formation in the pathogenesis of cGVHD. We aimed to assess the influence of ECP on the formation of NETs in patients with cGVHD as a potential mechanism in this setting. METHODS Patients treated with ECP for cGVHD at the Rabin Medical Center were included in this study. Blood samples were obtained at three different time points: before starting an ECP cycle, at the end of the first day of treatment, and 24 h following the initiation of the ECP treatment cycle. Neutrophils were harvested from all blood samples. NET formation was assessed by measurement of NET-bound specific neutrophil elastase activity and by immunofluorescence staining. RESULTS Six patients (two females and four males) with cGVHD were included in the study. We observed a significant increase in NET formation among all six patients following ECP. Net-bound specific neutrophil elastase activity was elevated from a median value of 2.23 mU/mL (interquartile range [IQR] 2.06-2.47 mU/mL) at baseline to a median value of 13.06 mU/mL (IQR 10.27-15.97 mU/mL) immediately after the treatment and to a peak median value of 14.73 mU/mL (IQR 9.6-22.38 mU/mL) 24 h following the initiation of the ECP cycle. A qualitative assessment of NET formation using immunofluorescence staining has demonstrated markedly increased expression of citrullinated histone H3, a marker of NET formation, following ECP treatment. CONCLUSIONS Our preliminary data indicate that ECP induces NET formation among patients with cGVHD. The contribution of increased NET formation to the therapeutic effect of cGVHD should be further investigated.
Collapse
Affiliation(s)
- Idan Goldberg
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Internal Medicine F - Recanati, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Galit Granot
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Alona Telerman
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Shirly Partouche
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Tzippy Shochat
- Statistical Consulting Unit, Beilinson Hospital, Rabin Medical Centre, Petah Tikva, Israel
| | - Erez Halperin
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Anat Gafter-Gvili
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Internal Medicine A, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
| | - Liat Shargian
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Yeshurun
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofir Wolach
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vered Yahalom
- Sackler School Medicine, Tel Aviv University, Tel Aviv, Israel
- Blood Services & Apheresis Institute, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
15
|
Huang F, Lv Y, Liu S, Wu H, Liu Q. Animal models for anti-neutrophil cytoplasmic antibody-associated vasculitis: Are current models good enough? Animal Model Exp Med 2023; 6:452-463. [PMID: 37614099 PMCID: PMC10614129 DOI: 10.1002/ame2.12345] [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: 04/03/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023] Open
Abstract
Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) is a rare and severe systemic autoimmune disease characterized by pauci-immune necrotizing inflammation of small blood vessels. AAV involves multiple organ systems throughout the body. Our knowledge of the pathogenesis of AAV has increased considerably in recent years, involving cellular, molecular and genetic factors. Because of the controlled environment with no other confounding factors, animal models are beneficial for studying the mechanistic details of disease development and for providing novel therapeutic targets with fewer toxic side effects. However, the complexity and heterogeneity of AAV make it very difficult to establish a single animal model that can fully represent the entire clinical spectrum found in patients. The aim of this review is to overview the current status of animal models for AAV, outline the pros and cons of methods, and propose potential directions for future research.
Collapse
Affiliation(s)
- Fei Huang
- Department of GeriatricsTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of General MedicineTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Yongman Lv
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of health management centerTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Siyang Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Wu
- Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Qingquan Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
16
|
Yadav R, Li QZ, Huang H, Bridges SL, Kahlenberg JM, Stecenko AA, Rada B. Cystic fibrosis autoantibody signatures associate with Staphylococcus aureus lung infection or cystic fibrosis-related diabetes. Front Immunol 2023; 14:1151422. [PMID: 37767091 PMCID: PMC10519797 DOI: 10.3389/fimmu.2023.1151422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction While cystic fibrosis (CF) lung disease is characterized by persistent inflammation and infections and chronic inflammatory diseases are often accompanied by autoimmunity, autoimmune reactivity in CF has not been studied in depth. Methods In this work we undertook an unbiased approach to explore the systemic autoantibody repertoire in CF using autoantibody microarrays. Results and discussion Our results show higher levels of several new autoantibodies in the blood of people with CF (PwCF) compared to control subjects. Some of these are IgA autoantibodies targeting neutrophil components or autoantigens linked to neutrophil-mediated tissue damage in CF. We also found that people with CF with higher systemic IgM autoantibody levels have lower prevalence of S. aureus infection. On the other hand, IgM autoantibody levels in S. aureus-infected PwCF correlate with lung disease severity. Diabetic PwCF have significantly higher levels of IgA autoantibodies in their circulation compared to nondiabetic PwCF and several of their IgM autoantibodies associate with worse lung disease. In contrast, in nondiabetic PwCF blood levels of IgA autoantibodies correlate with lung disease. We have also identified other autoantibodies in CF that associate with P. aeruginosa airway infection. In summary, we have identified several new autoantibodies and associations of autoantibody signatures with specific clinical features in CF.
Collapse
Affiliation(s)
- Ruchi Yadav
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA, United States
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hanwen Huang
- Department of Epidemiology & Biostatistics, College of Public Health, The University of Georgia, Athens, GA, United States
| | - S. Louis Bridges
- Department of Medicine, Hospital for Special Surgery, Division of Rheumatology, Weill Cornell Medical College, New York, NY, United States
| | - J. Michelle Kahlenberg
- Division of Rheumatology, University of Michigan, School of Medicine, Ann Arbor, MI, United States
| | - Arlene A. Stecenko
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA, United States
| |
Collapse
|
17
|
Qiao J, Zhang SX, Chang MJ, Zhao R, Song S, Hao JW, Wang C, Hu JX, Gao C, Wang CH, Li XF. Deep stratification by transcriptome molecular characters for precision treatment of patients with systemic lupus erythematosus. Rheumatology (Oxford) 2023; 62:2574-2584. [PMID: 36308437 DOI: 10.1093/rheumatology/keac625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/18/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES To leverage the high clinical heterogeneity of systemic lupus erythematosus (SLE), we developed and validated a new stratification scheme by integrating genome-scale transcriptomic profiles to identify patient subtypes sharing similar transcriptomic markers and drug targets. METHODS A normalized compendium of transcription profiles was created from peripheral blood mononuclear cells (PBMCs) of 1046 SLE patients and 86 healthy controls (HCs), covering an intersection of 13 689 genes from six microarray datasets. Upregulated differentially expressed genes were subjected to functional and network analysis in which samples were grouped using unsupervised clustering to identify patient subtypes. Then, clustering stability was evaluated by the stratification of six integrated RNA-sequencing datasets using the same method. Finally, the Xgboost classifier was applied to the independent datasets to identify factors associated with treatment outcomes. RESULTS Based on 278 upregulated DEGs of the transcript profiles, SLE patients were classified into three subtypes (subtype A-C) each with distinct molecular and cellular signatures. Neutrophil activation-related pathways were markedly activated in subtype A (named NE-driving), whereas lymphocyte and IFN-related pathways were more enriched in subtype B (IFN-driving). As the most severe subtype, subtype C [NE-IFN-dual-driving (Dual-driving)] shared functional mechanisms with both NE-driving and IFN-driving, which was closely associated with clinical features and could be used to predict the responses of treatment. CONCLUSION We developed the largest cohesive SLE transcriptomic compendium for deep stratification using the most comprehensive microarray and RNA sequencing datasets to date. This result could guide future design of molecular diagnosis and the development of stratified therapy for SLE patients.
Collapse
Affiliation(s)
- Jun Qiao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Min-Jing Chang
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Shan Song
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jia-Wei Hao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Can Wang
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Jing-Xi Hu
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Cai-Hong Wang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Xiao-Feng Li
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Limberg MM, Weihrauch T, Gray N, Ernst N, Hartmann K, Raap U. Eosinophils, Basophils, and Neutrophils in Bullous Pemphigoid. Biomolecules 2023; 13:1019. [PMID: 37509055 PMCID: PMC10377006 DOI: 10.3390/biom13071019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune blistering skin disease, of which the incidence has increased in recent years. BP is characterized by circulating IgG and IgE autoantibodies against the hemidesmosomal proteins BP180 and BP230. Although autoantibodies trigger inflammatory cascades that lead to blister formation, effector cells and cell-mediated autoimmunity must also be considered as important factors in the pathogenesis of BP. The aim of this review is to outline the current knowledge on the role of eosinophils, basophils, and neutrophils in BP.
Collapse
Affiliation(s)
- Maren M Limberg
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Tobias Weihrauch
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Natalie Gray
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Nancy Ernst
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Karin Hartmann
- Division of Allergy, Departments of Dermatology and Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Ulrike Raap
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- University Clinic of Dermatology and Allergy, University of Oldenburg, 26133 Oldenburg, Germany
| |
Collapse
|
19
|
Li C, Hu J, Xing Y, Han J, Zhang A, Zhang Y, Hua Y, Tian Z, Bai Y. Constraint-induced movement therapy alleviates motor impairment by inhibiting the accumulation of neutrophil extracellular traps in ischemic cortex. Neurobiol Dis 2023; 179:106064. [PMID: 36878327 DOI: 10.1016/j.nbd.2023.106064] [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: 10/11/2022] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Stroke is a major cause of mortality and morbidity and most acute strokes are ischemic. Evidence-based medicine has demonstrated the effectiveness of constraint-induced movement therapy (CIMT) in the recovery of motor function in patients after ischemic stroke, but the specific treatment mechanism remains unclear. Herein, our integrated transcriptomics and multiple enrichment analysis studies, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) studies show that CIMT conduction broadly curtails immune response, neutrophil chemotaxis, and chemokine-mediated signaling pathway, CCR chemokine receptor binding. Those suggest the potential effect of CIMT on neutrophils in ischemic mice brain parenchyma. Recent studies have found that accumulating granulocytes release extracellular web-like structures composed of DNA and proteins called neutrophil extracellular traps (NETs), which destruct neurological function primarily by disrupting the blood-brain barrier and promoting thrombosis. However, the temporal and spatial distribution of neutrophils and their released NETs in parenchyma and their damaging effects on nerve cells remain unclear. Thus, utilizing immunofluorescence and flow cytometry, our analyses uncovered that NETs erode multiple regions such as primary motor cortex (M1), striatum (Str), nucleus of the vertical limb of the diagonal band (VDB), nucleus of the horizontal limb of the diagonal band (HDB) and medial septal nucleus (MS), and persist in the brain parenchyma for at least 14 days, while CIMT can reduce the content of NETs and chemokines CCL2 and CCL5 in M1. Intriguingly, CIMT failed to further reduce neurological deficits after inhibiting the NET formation by pharmacologic inhibition of peptidylarginine deiminase 4 (PAD4). Collectively, these results demonstrate that CIMT could alleviate cerebral ischemic injury induced locomotor deficits by modulating the activation of neutrophils. These data are expected to provide direct evidence for the expression of NETs in ischemic brain parenchyma and novel insights into the mechanisms of CIMT protecting against ischemic brain injury.
Collapse
Affiliation(s)
- Congqin Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Xing
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Han
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Brain Science Collaborative Innovation Center, School of Basic Medical Sciences, Institutes of Brain Science, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Anjing Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuqian Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhanzhuang Tian
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Brain Science Collaborative Innovation Center, School of Basic Medical Sciences, Institutes of Brain Science, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
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: 2.5] [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
|
21
|
Necrostatin-1 Alleviates Diffuse Pulmonary Haemorrhage by Preventing the Release of NETs via Inhibiting NE/GSDMD Activation in Murine Lupus. J Immunol Res 2023; 2023:4743975. [PMID: 36910905 PMCID: PMC9995194 DOI: 10.1155/2023/4743975] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 03/06/2023] Open
Abstract
Diffuse alveolar haemorrhage (DAH) is a rapidly developing condition owing to a lack of effective treatment and resulting in a high mortality rate in systemic lupus erythematosus (SLE). Neutrophil extracellular traps (NETs) contain numerous antigens and proinflammatory substances that directly damage the vascular endothelium and aggravate vascular inflammation, which is considered an important pathogenic factor of DAH in SLE. Therefore, blocking the release of NETs from neutrophils is an important target for the treatment of DAH in SLE. In this study, we investigated whether the inhibition of neutrophils releasing NETs could relieve DAH in SLE. Necrostatin-1 (Nec-1), a small molecule, has been reported to inhibit the release of NETs by neutrophils. In vitro experiments revealed that Nec-1 inhibited alveolar epithelial cell damage by preventing the release of NETs. Furthermore, vivo studies showed that Nec-1 alleviated lupus pulmonary haemorrhage in mice by reducing lung pathology severity, body weight, and serum inflammatory cytokine levels. Mechanistically, Nec-1 prevented NET release by inhibiting neutrophil elastase (NE) activation and N-Gasdermin D (N-GSDMD) expression. Additionally, immunohistochemistry and immunofluorescence findings showed that Nec-1 decreased NE expression in the lung tissues of mice with lupus pulmonary haemorrhage. Thus, NETs released by neutrophils contributed to the pathogenesis of DAH in SLE, and Nec-1 showed protective effects by the inhibition of NET production via the reduction of NE activation and N-GSDMD expression.
Collapse
|
22
|
Role of neutrophil extracellular traps in inflammatory evolution in severe acute pancreatitis. Chin Med J (Engl) 2022; 135:2773-2784. [PMID: 36729096 PMCID: PMC9945416 DOI: 10.1097/cm9.0000000000002359] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Indexed: 02/03/2023] Open
Abstract
ABSTRACT Severe acute pancreatitis (SAP) is a life-threatening acute abdominal disease with two peaks of death: the first in the early stage, characterized by systemic inflammatory response-associated organ failure; and the second in the late stage, characterized by infectious complications. Neutrophils are the main immune cells participating in the whole process of SAP. In addition to the traditional recognition of neutrophils as the origination of chemokine and cytokine cascades or phagocytosis and degranulation of pathogens, neutrophil extracellular traps (NETs) also play an important roles in inflammatory reactions. We reviewed the role of NETs in the occurrence and development of SAP and its fatal complications, including multiple organs injury, infected pancreatic necrosis, and thrombosis. This review provides novel insights into the involvement of NETs throughout the entire process of SAP, showing that targeting NETs might be a promising strategy in SAP treatment. However, precision therapeutic options targeting NETs in different situations require further investigation.
Collapse
|
23
|
Ni S, Yuan Y, Song S, Li X. A double-edged sword with a therapeutic target: iron and ferroptosis in immune regulation. Nutr Rev 2022; 81:587-596. [PMID: 36130411 DOI: 10.1093/nutrit/nuac071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cellular activities such as DNA synthesis, adenosine triphosphate production, and mitochondrial respiration are affected by iron metabolism. Disturbance of iron homeostasis usually leads to damage in cells and organs in the context of iron overload or deficiency. Thus, iron, a key regulator in nutritional immunity, was shown to be critical in innate and adaptive immunity. Unlike apoptosis, ferroptosis, a feature of iron-dependent lipid peroxidation, is thought to be associated with immune regulation because of its immunogenic nature. In this review, we summarize the role of iron and ferroptosis in immune regulation and discuss their therapeutic potential in the treatment of arthropathies like osteoarthritis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Shuo Ni
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yin Yuan
- the State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shangdao Song
- the Department of Rehabilitation Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiaolin Li
- the Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
24
|
Klas K, Ondracek AS, Hofbauer TM, Mangold A, Pfisterer K, Laggner M, Copic D, Direder M, Bormann D, Ankersmit HJ, Mildner M. The Effect of Paracrine Factors Released by Irradiated Peripheral Blood Mononuclear Cells on Neutrophil Extracellular Trap Formation. Antioxidants (Basel) 2022; 11:antiox11081559. [PMID: 36009277 PMCID: PMC9405389 DOI: 10.3390/antiox11081559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Neutrophil extracellular trap (NET)-formation represents an important defence mechanism for the rapid clearance of infections. However, exaggerated NET formation has been shown to negatively affect tissue-regeneration after injury. As our previous studies revealed the strong tissue-protective and regenerative properties of the secretome of stressed peripheral blood mononuclear cells (PBMCsec), we here investigated the influence of PBMCsec on the formation of NETs. The effect of PBMCsec on NET formation was assessed ex vivo in ionomycin stimulated neutrophils derived from healthy donors using flow cytometry, image stream analysis, and quantification of released extracellular DNA. The effect of PBMCsec on molecular mechanisms involved in NET formation, including Ca-flux, protein kinase C activity, reactive oxygen species production, and protein arginine deiminase 4 activity, were analysed. Our results showed that PBMCsec significantly inhibited NET formation. Investigation of the different biological substance classes found in PBMCsec revealed only a partial reduction in NET formation, suggesting a synergistic effect. Mechanistically, PBMCsec treatment did not interfere with calcium signalling and PKC-activation, but exerted anti-oxidant activity, as evidenced by reduced levels of reactive oxygen species and upregulation of heme oxygenase 1 and hypoxia inducible-factor 1 in PBMCsec-treated neutrophils. In addition, PBMCsec strongly inhibited the activation of protein arginine deiminase 4 (PAD4), ultimately leading to the inhibition of NET formation. As therapeutics antagonizing excessive NET formation are not currently available, our study provides a promising novel treatment option for a variety of conditions resulting from exaggerated NET formation.
Collapse
Affiliation(s)
- Katharina Klas
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Anna S Ondracek
- Division of Cardiology, Department of Internal Medicine II, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas M Hofbauer
- Division of Cardiology, Department of Internal Medicine II, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Mangold
- Division of Cardiology, Department of Internal Medicine II, Vienna General Hospital, Medical University of Vienna, 1090 Vienna, Austria
| | - Karin Pfisterer
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Dragan Copic
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Martin Direder
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Daniel Bormann
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Hendrik Jan Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
25
|
Li H, Yang P. Identification of biomarkers related to neutrophils and two molecular subtypes of systemic lupus erythematosus. BMC Med Genomics 2022; 15:162. [PMID: 35858908 PMCID: PMC9297641 DOI: 10.1186/s12920-022-01306-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE), an autoimmune disease with complex pathogenesis, poses a considerable threat to women’s health. Increasing evidence indicates that neutrophils play an important role in the development and progression of lupus. Methods Weighted correlation network analysis and single-sample gene set enrichment analysis (GSEA) were used to analyse SLE expression data from a comprehensive gene expression database and identify modules associated with neutrophils. Thereafter, the biomarkers most closely related to neutrophils were identified. We reclassified SLE into two molecular subtypes based on the aforementioned biomarkers and evaluated cell infiltration, molecular mechanisms, and signature pathways in each subtype. Results The results showed significant differences in immunological characteristics between the two molecular subtypes of SLE. Hub genes were significantly upregulated in the NEUT-H subtype, and they may be associated with lupus activity. The GSEA revealed associations between our biomarkers and key metabolic pathways. Conclusions Our study provides not only a classification for patients with SLE but also new cell and gene targets for immunotherapy, as well as a new experimental paradigm to explore immunotherapy for other autoimmune diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01306-9.
Collapse
Affiliation(s)
- Huiyan Li
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, 110001, China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, 110001, China.
| |
Collapse
|
26
|
Pfister H. Neutrophil Extracellular Traps and Neutrophil-Derived Extracellular Vesicles: Common Players in Neutrophil Effector Functions. Diagnostics (Basel) 2022; 12:diagnostics12071715. [PMID: 35885618 PMCID: PMC9323717 DOI: 10.3390/diagnostics12071715] [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: 06/02/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Neutrophil granulocytes are a central component of the innate immune system. In recent years, they have gained considerable attention due to newly discovered biological effector functions and their involvement in various pathological conditions. They have been shown to trigger mechanisms that can either promote or inhibit the development of autoimmunity, thrombosis, and cancer. One mechanism for their modulatory effect is the release of extracellular vesicles (EVs), that trigger appropriate signaling pathways in immune cells and other target cells. In addition, activated neutrophils can release bactericidal DNA fibers decorated with proteins from neutrophil granules (neutrophil extracellular traps, NETs). While NETs are very effective in limiting pathogens, they can also cause severe damage if released in excess or cleared inefficiently. Since NETs and EVs share a variety of neutrophil molecules and initially act in the same microenvironment, differential biochemical and functional analysis is particularly challenging. This review focuses on the biochemical and functional parallels and the extent to which the overlapping spectrum of effector molecules has an impact on biological and pathological effects.
Collapse
Affiliation(s)
- Heiko Pfister
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Center Munich, Technical University Munich, D-80636 Munich, Germany
| |
Collapse
|
27
|
Choi Y, Luu QQ, Park HS. Extracellular Traps: A Novel Therapeutic Target for Severe Asthma. J Asthma Allergy 2022; 15:803-810. [PMID: 35726304 PMCID: PMC9206515 DOI: 10.2147/jaa.s366014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/24/2022] [Indexed: 01/18/2023] Open
Abstract
Asthma is a complicated disease defined by a combination of clinical symptoms and physiological characteristics. Typically, asthma is diagnosed by the presence of episodic cough, wheezing, or dyspnea triggered by variable environmental factors (allergens and respiratory infections), and reversible airflow obstruction. To date, the majority of asthmatic patients have been adequately controlled by anti-inflammatory/bronchodilating agents, but those with severe asthma (SA) have not been sufficiently controlled by high-dose inhaled corticosteroids-long-acting beta-agonists plus additional controllers including leukotriene modifiers. Accordingly, these uncontrolled patients provoke a special issue, because they consume high healthcare resources, requiring innovative precision medicine solutions. Recently, phenotyping based on biomarkers of airway inflammation has led to elucidating the pathophysiological mechanism of SA, where emerging evidence has highlighted the significance of eosinophil or neutrophil extracellular traps contributing to the development of SA. Here, we aimed to provide current findings about extracellular traps as a novel therapeutic target for asthma to address medical unmet needs.
Collapse
Affiliation(s)
- Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Quoc Quang Luu
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Korea
| |
Collapse
|
28
|
Chen XQ, Tu L, Tang Q, Huang L, Qin YH. An Emerging Role for Neutrophil Extracellular Traps in IgA Vasculitis: A Mini-Review. Front Immunol 2022; 13:912929. [PMID: 35799774 PMCID: PMC9253285 DOI: 10.3389/fimmu.2022.912929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Immunoglobulin A vasculitis (IgAV) is the most common systemic small vessel vasculitis in childhood. Its clinical manifestations are non-thrombocytopenic purpura, accompanied by gastrointestinal tract, joint, kidney and other organ system involvement. The pathogenesis of IgAV has not been fully elucidated. It may be related to many factors including genetics, infection, environmental factors, and drugs. The most commonly accepted view is that galactose-deficient IgA1 and the deposition of IgA and complement C3 in small blood vessel walls are key contributors to the IgAV pathogenesis. Extensive neutrophil extracellular traps (NETs) in the peripheral circulation and skin, kidney, and gastrointestinal tissue of patients with IgAV has been identified in the past two years and is associated with disease activity. This mini-review provides a possible mechanism for NETs involvement in the pathogenesis of IgAV.
Collapse
|
29
|
Tu J, Jin J, Chen X, Sun L, Cai Z. Altered Cellular Immunity and Differentially Expressed Immune-Related Genes in Patients With Systemic Sclerosis-Associated Pulmonary Arterial Hypertension. Front Immunol 2022; 13:868983. [PMID: 35663995 PMCID: PMC9159786 DOI: 10.3389/fimmu.2022.868983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is the most common connective tissue disease causing pulmonary hypertension (PAH). However, the cause and potential immune molecular events associated with PAH are still unclear. Therefore, it is particularly essential to analyze the changes in SSc-PAH–related immune cells and their immune-related genes. Three microarray datasets (GSE22356, GSE33463, and GSE19617) were obtained by the Gene Expression Omnibus (GEO). Compared with SSc, we found neutrophils have a statistically higher abundance, while T-cell CD4 naive and T-cell CD4 memory resting have a statistically lower abundance in peripheral blood mononuclear cells (PBMCs). Moreover, the results of Gene Set Enrichment Analysis (GSEA) showed there is a differential enrichment of multiple pathways between SSc and SSc-PAH. By combining differentiated expressed genes (DEGs) and immune-related genes (IRGs), fifteen IRGs were selected. In addition, we also analyzed the first five rich Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and the most abundant Gene Ontology (GO)-molecular functional terms. Furthermore, interleukin-7 receptor (IL-7R), tyrosine–protein kinase (LCK), histone deacetylase 1 (HDAC1), and epidermal growth factor receptor (EGFR) genes were identified as hub genes via protein–protein interaction (PPI) network analysis. The Comparative Toxic Genomics Database (CTD) analysis result showed that LCK, HDAC1, and EGFR have a higher score with SSc. Coexpression network analysis confirmed that IL-7R, LCK, and HDAC1 are key genes related to immune regulation in SSc without PAH and are involved in T-cell immune regulation. Subsequently, using GSE22356 and GSE33463 as the test sets and GSE19617 as the verification set, it was verified that the mRNA expression levels of the three central genes of SSc-PAH were significantly lower than those of the SSc without PAH samples. Consistent with previous predictions, the expressions of IL-7R, LCK, and HDAC1 are positively correlated with the numbers of T-cell CD4 naive and T-cell CD4 memory, while the expressions of IL-7R and LCK are negatively correlated with the numbers of neutrophils in the peripheral blood. Therefore, this evidence may suggest that these three immune-related genes: IL-7R, LCK, and HDAC1, may be highly related to the immunological changes in SSc-PAH. These three molecules can reduce T cells in SSc-PAH PBMCs through the regulation of T-cell activation, which suggests that these three molecules may be involved in the development of SSc-PAH. Meanwhile, the low expression of IL-7R, LCK, and HDAC1 detected in the peripheral blood of SSc may indicate the possibility of PAH and hopefully become a biomarker for the early detection of SSc-PAH. Finally, 49 target miRNAs of 3 specifically expressed hub genes were obtained, and 49 mRNA–miRNA pairs were identified, which provided directions for our further research.
Collapse
Affiliation(s)
- Jianxin Tu
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Rheumatology, The First Affiliated Hospital of Wenzhou University, Wenzhou, China
| | - Jinji Jin
- Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou University, Wenzhou, China
| | - Xiaowei Chen
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou University, Wenzhou, China
| | - Li Sun
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou University, Wenzhou, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
30
|
Zhou Y, Xu Z, Liu Z. Impact of Neutrophil Extracellular Traps on Thrombosis Formation: New Findings and Future Perspective. Front Cell Infect Microbiol 2022; 12:910908. [PMID: 35711663 PMCID: PMC9195303 DOI: 10.3389/fcimb.2022.910908] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022] Open
Abstract
Thrombotic diseases seriously endanger human health, neutrophils and neutrophil extracellular traps (NETs) play an important role in abnormal thrombus formation. NETs are extracellular structures released by neutrophils upon stimulation by pathogens. NETs include neutrophil elastase (NE), myeloperoxidase (MPO), cathepsin G and other active substances. The network structure provided by NETs can prevent the spread of pathogens and effectively kill and eliminate pathogens. However, the components of NETs can also abnormally activate the coagulation pathway and participate in the formation of pathological thrombi. This review aims to summarize the mechanisms of NETs formation in detail; the research progress of NETs in venous thrombosis, arterial thrombosis, acquired disease-associated thrombosis, sepsis coagulation disorder; as well as the strategies to target NETs in thrombosis prevention and treatment.
Collapse
Affiliation(s)
| | - Zhendong Xu
- *Correspondence: Zhiqiang Liu, ; Zhendong Xu,
| | | |
Collapse
|
31
|
Keir HR, Chalmers JD. Neutrophil extracellular traps in chronic lung disease: implications for pathogenesis and therapy. Eur Respir Rev 2022; 31:31/163/210241. [PMID: 35197267 PMCID: PMC9488971 DOI: 10.1183/16000617.0241-2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/28/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophilic inflammation has a key role in the pathophysiology of multiple chronic lung diseases. The formation of neutrophil extracellular traps (NETs) has emerged as a key mechanism of disease in neutrophilic lung diseases including asthma, COPD, cystic fibrosis and, most recently, bronchiectasis. NETs are large, web-like structures composed of DNA and anti-microbial proteins that are able to bind pathogens, prevent microbial dissemination and degrade bacterial virulence factors. The release of excess concentrations of proteases, antimicrobial proteins, DNA and histones, however, also leads to tissue damage, impaired mucociliary clearance, impaired bacterial killing and increased inflammation. A number of studies have linked airway NET formation with greater disease severity, increased exacerbations and overall worse disease outcomes across the spectrum of airway diseases. Treating neutrophilic inflammation has been challenging in chronic lung disease because of the delicate balance between reducing inflammation and increasing the risk of infections through immunosuppression. Novel approaches to suppressing NET formation or the associated inflammation are in development and represent an important therapeutic target. This review will discuss the relationship between NETs and the pathophysiology of cystic fibrosis, asthma, COPD and bronchiectasis, and explore the current and future development of NET-targeting therapies. NETs contribute to the pathophysiology of chronic lung disease. Immunomodulating therapies that may reduce inflammatory mediators and NET formation, without compromising bacterial clearance, offer a new treatment path for patients. https://bit.ly/3fyJC6I
Collapse
Affiliation(s)
- Holly R Keir
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
32
|
Bolouri N, Akhtari M, Farhadi E, Mansouri R, Faezi ST, Jamshidi A, Mahmoudi M. Role of the innate and adaptive immune responses in the pathogenesis of systemic lupus erythematosus. Inflamm Res 2022; 71:537-554. [PMID: 35298669 DOI: 10.1007/s00011-022-01554-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE), the most common form of lupus, is a multisystemic rheumatic disease with different clinical features that generally affect women of childbearing age. The common symptoms of SLE are very similar to other autoimmune and non-autoimmune disorders, thereby it is known as a thousand faces disease. In this article, we are going to discuss some of the most updated information about immune system-related factors, cells, and cytokines involved in SLE pathogenesis. METHODS Different electronic databases, especially PubMed/MEDLINE, Scopus, and Google Scholar, were searched to review and analyze relevant literature on the role of innate and adaptive immune cells and cytokines in the pathogenesis of SLE. A search for relevant literature was accomplished using various keywords including systemic lupus erythematosus, apoptosis, autoantibodies, immunopathogenesis of SLE, adaptive and innate immune cells, inflammatory cytokines, hormones, etc. RESULTS AND CONCLUSION: The most important characteristic of SLE is the production of antibodies against different nuclear autoantigens like double-strand DNA and RNA. The depositions of the immune complexes (ICs) that are generated between autoantibodies and autoantigens, along with aberrant clearance of them, can lead to permanent inflammation and contribute to tissue or organ damage. Related mechanisms underlying the initiation and development of SLE have not been clarified yet. Although, defects in immune tolerance, enhanced antigenic load, hyperactivity of T cells, and inappropriate regulation of B cells contribute to the pathogenic autoantibodies generation. Besides, sex hormones that influence the immune system seem to act as triggers or protectors of SLE development.
Collapse
Affiliation(s)
- Nasim Bolouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Akhtari
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. .,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Mansouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedeh Tahereh Faezi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Torres-Ruiz J, Carrillo-Vázquez DA, Leal-Alanis A, Zentella-Dehesa A, Tapia-Rodríguez M, Maravillas-Montero JL, Nuñez-Álvarez CA, Carazo-Vargas ER, Romero-Hernández I, Juárez-Vega G, Alcocer-Varela J, Gómez-Martín D. Low-Density Granulocytes and Neutrophil Extracellular Traps as Biomarkers of Disease Activity in Adult Inflammatory Myopathies. J Clin Rheumatol 2022; 28:e480-e487. [PMID: 34643846 DOI: 10.1097/rhu.0000000000001772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND/OBJECTIVE Biomarkers for disease activity and damage accrual in idiopathic inflammatory myopathies (IIMs) are currently lacking. The purpose of this cross-sectional study is to analyze the relationship among low-density granulocytes (LDGs), neutrophil extracellular traps (NETs), and clinical and immunological features of patients with IIM. METHODS We assessed disease activity, damage accrual, amount of LDGs, NETs, expression of LL-37, and serum cytokines in 65 adult patients with IIM. Differences between groups and correlations were assessed by Kruskal-Wallis, Mann-Whitney U, and Spearman ρ tests. The association between LDGs, NETs, disease activity, calcinosis, and cutaneous ulcers was assessed by logistic regression. To address the capacity of LDGs and NETs to diagnose disease activity, we used receiving operating characteristic curves. RESULTS Low-density granulocytes were higher in patients with active disease, ulcers, calcinosis, and anti-MDA5 antibodies, which correlated with serum levels of IL-17A and IL-18. Neutrophil extracellular traps were higher in patients with calcinosis, elevated titers of antinuclear antibodies, and positive anti-PM/Scl75 tests. The combination of a high proportion of both total LDGs and NETs was associated with the presence of calcinosis and cutaneous ulcers. LL-37 was higher in NETs originating from LDGs. Normal-density neutrophils were elevated in patients with active dermatomyositis. CONCLUSIONS Low-density granulocytes and NETs containing LL-37 are increased in patients with IIM and active disease, and correlate with proinflammatory cytokines. Both total and CD10+ LDGs are potential biomarkers for disease activity and, in combination with NETs, have the potential to detect patients who are at risk for cutaneous ulcers and calcinosis.
Collapse
Affiliation(s)
| | | | - Araceli Leal-Alanis
- Internal Medicine, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran
| | | | - Miguel Tapia-Rodríguez
- Microscopy Unit, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico
| | | | | | | | | | - Guillermo Juárez-Vega
- Flow Cytometry Unit, Red de Apoyo a la Investigación, Coordinacion de Investigación Cientifica, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | | | | |
Collapse
|
34
|
Kim Y, Cho AY, Kim HC, Ryu D, Jo SA, Jung YS. Effects of Natural Polyphenols on Oxidative Stress-Mediated Blood–Brain Barrier Dysfunction. Antioxidants (Basel) 2022; 11:antiox11020197. [PMID: 35204080 PMCID: PMC8868362 DOI: 10.3390/antiox11020197] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
The blood-brain barrier (BBB), which consists mainly of brain microvascular endothelial cells and astrocytes connected by tight junctions (TJs) and adhesion molecules (AMs), maintains the homeostatic balance between brain parenchyma and extracellular fluid. Accumulating evidence shows that BBB dysfunction is a common feature of neurodegenerative diseases, including stroke, traumatic brain injury, and Alzheimer’s disease. Among the various pathological pathways of BBB dysfunction, reactive oxygen species (ROS) are known to play a key role in inducing BBB disruption mediated via TJ modification, AM induction, cytoskeletal reorganization, and matrix metalloproteinase activation. Thus, antioxidants have been suggested to exert beneficial effects on BBB dysfunction-associated brain diseases. In this review, we summarized the sources of ROS production in multiple cells that constitute or surround the BBB, such as BBB endothelial cells, astrocytes, microglia, and neutrophils. We also reviewed various pathological mechanisms by which BBB disruption is caused by ROS in these cells. Finally, we summarized the effects of various natural polyphenols on BBB dysfunction to suggest a therapeutic strategy for BBB disruption-related brain diseases.
Collapse
Affiliation(s)
- Yeonjae Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - A Yeon Cho
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Hong Cheol Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Dajung Ryu
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea;
- Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan 31116, Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
35
|
Misra DP, Thomas KN, Gasparyan AY, Zimba O. Mechanisms of thrombosis in ANCA-associated vasculitis. Clin Rheumatol 2021; 40:4807-4815. [PMID: 34109491 PMCID: PMC8189705 DOI: 10.1007/s10067-021-05790-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/07/2021] [Accepted: 05/22/2021] [Indexed: 12/19/2022]
Abstract
Patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) have a two- to threefold greater risk of developing venous as well as arterial thrombotic events. Although such thrombotic events are more commonly seen during phases of active AAV, they are also recognized to occur during AAV in remission. Endothelial injury is a key pathogenic event in AAV. Endothelial injury can be caused by neutrophil activation and release of thrombogenic tissue factor into the circulation. Neutrophil activation further results in the formation of neutrophil extracellular traps (NETs). NETs contribute to thrombosis by expressing tissue factor. NETs have also been detected in cutaneous thrombi from patients with AAV induced by hydralazine. Activated neutrophils in AAV patients release thrombogenic microparticles loaded with tissue factor which further enhances clotting of blood. Antiphospholipid antibodies (APLs) have been detected in up to a third of AAV and might also be induced by drugs such as cocaine adulterated with levamisole and propylthiouracil, which are known to trigger AAV. Such APLs further drive the thrombosis in AAV. Once thrombogenesis occurs, the homeostatic mechanisms resulting in clot dissolution are further impaired in AAV due to anti-plasminogen antibodies. The ongoing pandemic of coronavirus disease 2019 (COVID-19) is associated with endothelial injury and NETosis, mechanisms which are in common with AAV. Reports of new-onset AAV following COVID-19 have been described in the literature, and there could be shared mechanisms driving these processes that require further evaluation.
Collapse
Affiliation(s)
- Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India.
| | - Koshy Nithin Thomas
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands, UK
| | - Olena Zimba
- Department of Internal Medicine #2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
36
|
Cody EM, Brunner HI. Biomarkers in Childhood-Onset Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 48:271-285. [PMID: 34798952 DOI: 10.1016/j.rdc.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex, multisystem chronic autoimmune disease. Because of its diverse phenotypes, diagnosis of SLE can be challenging, and current biomarkers are insufficient. Childhood-onset SLE (cSLE), although less prevalent, has higher morbidity and mortality, and early diagnosis is critical for improving outcomes. Many studies have focused on discovering new biomarkers to better diagnose and monitor SLE and cSLE. Herein, the authors aim to review the most investigated biomarkers in development for cSLE, focusing on those that can be measured in the blood or urine.
Collapse
Affiliation(s)
- Ellen M Cody
- Division of Pediatric Nephrology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MC 7022, Cincinnati, OH 45229-3930, USA.
| | - Hermine I Brunner
- Division of Rheumatology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MC 4010, Cincinnati, OH 45229-3930, USA
| |
Collapse
|
37
|
Neutrophil Extracellular Traps Exacerbate Ischemic Brain Damage. Mol Neurobiol 2021; 59:643-656. [PMID: 34748205 DOI: 10.1007/s12035-021-02635-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
Most acute strokes are ischemic, and subsequent neuroinflammation promotes further damage leading to cell death but also plays a beneficial role by promoting cellular repair. Neutrophils are forerunners to brain lesions after ischemic stroke and exert elaborate functions. While neutrophil extracellular traps (NETs) possess a fundamental antimicrobial function within the innate immune system under physiological circumstances, increasing evidence indicates that NETosis, the release process of NETs, occurs in the pathogenic process of stroke. In this review, we focus on the processes of NET formation and clearance, the temporal and spatial alterations of neutrophils and NETs after ischemic damage, and how NETs are involved in several stroke-related phenomena. Generally, NET formation and release processes depend on the generation of reactive oxygen species (ROS) and the activation of nuclear peptidylarginine deiminase-4 (PAD4). The acid-base environment, oxygen concentration, and iron ions around the infarct may also impact NET formation. DNase 1 has been identified as the primary degrader of NETs in serum, while reactive microglia are expected to inhibit the formation of NETs around ischemic lesions by phagocytosis of neutrophils. The neutrophils and NETs are present in the perivascular space ipsilateral to the infarct arising after ischemic damage, peaking between 1 and 3 days postischemia, but their location in the brain parenchyma remains controversial. After the ischemic injury, NETs are involved in the destruction of neurological function primarily by disrupting the blood-brain barrier and promoting thrombosis. The potential effects of NETs on various ischemic nerve cells need to be further investigated, especially in the chronic ischemic phase.
Collapse
|
38
|
Shen D, Lu Y, Li G, Hu M, Li S, Ju H, Zhang M, Wang X. Mechanism of neutrophil extracellular traps generation and their role in trophoblasts apoptosis in gestational diabetes mellitus. Cell Signal 2021; 88:110168. [PMID: 34634453 DOI: 10.1016/j.cellsig.2021.110168] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 12/23/2022]
Abstract
Gestational diabetes mellitus (GDM) is a metabolic syndrome occurring in pregnant women and increases the risk of placental dysplasia. Neutrophil extracellular traps (NETs) may play a critical role in placental dysplasia. NETosis (neutrophil cell death by NET release) depends on NADPH/ROS pathway. In view of the adiponectin which is widely believed to be reduced in GDM patients suppresses NADPH oxidase and ROS generation of neutrophil. We speculate that increased NET release is associated with hypoadiponectinemia. Trophoblast apoptosis is significantly increased in GDM patients, but it is not clear whether NETs promotes cell apoptosis. This study aims to reveal the mechanism of Neutrophil Extracellular Traps generation and their role in trophoblast apoptosis in Gestational Diabetes Mellitus. We investigated the generation of NETs by cell-free DNA (cf-DNA) quantification, live-cell imaging, and reactive oxygen species (ROS) measurement. ERK1/2 and p38 MAPK signalling pathway proteins were detected by western blotting. The Cell Counting Kit-8 (CCK-8) assay, flow cytometry, and western blotting were performed to explore the effects of NETs on trophoblast apoptosis. We found that adiponectin inhibited NET release by suppressing ROS production, and p38 MAPK and ERK1/2 proteins were involved in the process. Further, NETs promoted trophoblast apoptosis by activating the ROS-dependent mitochondrial pathway, which is mediated by ERK1/2 signalling. The current study demonstrated that hypoadiponectinemia is the cause of NETs formation and NETs promoting trophoblast apoptosis.
Collapse
Affiliation(s)
- Di Shen
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Street, Jinan, Shandong 250021, China; Department of Obstetrics and Gynaecology, Maternal and Child Health Care Hospital of Shandong Provincial, Cheeloo College of Medicine, Shandong University, 238 Jingshi East Road, Jinan, Shandong 250014, China
| | - Yuan Lu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Street, Jinan, Shandong 250021, China
| | - Guangzhen Li
- Department of General Surgery, Qilu Hospital of Shandong University, 107 Wenhua West Road, 251000, Jinan, China
| | - Min Hu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Street, Jinan, Shandong 250021, China
| | - Shanling Li
- Department of Obstetrics and Gynaecology, Maternal and Child Health Care Hospital of Shandong Provincial, Cheeloo College of Medicine, Shandong University, 238 Jingshi East Road, Jinan, Shandong 250014, China
| | - Hui Ju
- Department of Obstetrics and Gynaecology, Maternal and Child Health Care Hospital of Shandong Provincial, Cheeloo College of Medicine, Shandong University, 238 Jingshi East Road, Jinan, Shandong 250014, China
| | - Meihua Zhang
- The Laboratory of Placenta-Related Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, 238 Jingshi East Road, Jinan, Shandong 250014, China
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Street, Jinan, Shandong 250021, China; Department of Obstetrics and Gynaecology, Maternal and Child Health Care Hospital of Shandong Provincial, Cheeloo College of Medicine, Shandong University, 238 Jingshi East Road, Jinan, Shandong 250014, China; The Laboratory of Placenta-Related Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, 238 Jingshi East Road, Jinan, Shandong 250014, China.
| |
Collapse
|
39
|
Peptidylarginine deiminases 4 as a promising target in drug discovery. Eur J Med Chem 2021; 226:113840. [PMID: 34520958 DOI: 10.1016/j.ejmech.2021.113840] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/21/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Peptidylarginine deaminase 4 (PAD4) is a crucial post-translational modifying enzyme catalyzing the conversion of arginine into citrulline residues, and mediating the formation of neutrophil extracellular traps (NETs). PAD4 plays a vital role in the occurrence and development of cardiovascular diseases, autoimmune diseases, and various tumors. Therefore, PAD4 is considered as a promising drug target for disease diagnosis and treatment. More and more efforts are devoted to developing highly efficient and selective PAD4 inhibitors via high-throughput screening, structure-based drug design and structure-activity relationship study. This article outlined the physiological and pathological functions of PAD4, and corresponding representative small molecule inhibitors reported in recent years.
Collapse
|
40
|
Mozzini C, Pagani M. Clonal Hematopoiesis and Cardiovascular Diseases: The Connection. Curr Probl Cardiol 2021; 47:100962. [PMID: 34391764 DOI: 10.1016/j.cpcardiol.2021.100962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/08/2021] [Indexed: 01/22/2023]
Abstract
Atherothrombosis is the leading cause of death worldwide, but the precise mechanisms are not yet fully understood. Traditional cardiovascular risk factors have been known for many years, but are not enough to predict individual risk despite consolidated and emerging risk scoring systems. Clonal Haematopoiesis of Indeterminate Potential (CHIP) refers to the clonal expansion of a population of haematopoietic cells in response to the acquisition of a somatic mutation, without any clinical or biological sign of haematological malignancy. The prevalence of this condition increases with age, reaching 10 to20% of the general population aged >70 years. Recent studies have shown a link between CHIP and cardiovascular diseases. CHIP carriers have higher risk of cardiovascular diseases with also a more severe prognosis. Inflammation and immunity play a critical role in enhancing the cardiovascular consequences of CHIP. In this review we discuss the association between CHIP and cardiovascular diseases focusing on inflammation and other pathways shared with atherosclerosis progression. It is hopeful that in the future patients recognized as CHIP carriers may be classified as high-risk cardiovascular patients and new treatment targets will be required for them.
Collapse
Affiliation(s)
- Chiara Mozzini
- Department of Medicine, Section of Internal Medicine, Carlo Poma Hospital, Mantova Italy.
| | - Mauro Pagani
- Department of Medicine, Section of Internal Medicine, Carlo Poma Hospital, Mantova Italy
| |
Collapse
|
41
|
Fang H, Shao S, Xue K, Yuan X, Qiao P, Zhang J, Cao T, Luo Y, Bai X, Li W, Li C, Qiao H, Dang E, Wang G. Neutrophil extracellular traps contribute to immune dysregulation in bullous pemphigoid via inducing B-cell differentiation and antibody production. FASEB J 2021; 35:e21746. [PMID: 34151465 DOI: 10.1096/fj.202100145r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022]
Abstract
Bullous pemphigoid (BP), an autoimmune skin disease, is characterized by autoantibodies against hemidesmosomal proteins in the skin and mucous membranes. Neutrophils infiltrate BP skin lesions, however, their role in immune dysregulation remains unclear. We investigated whether BP involves aberrant neutrophil extracellular traps (NETs) formation in skin lesions and circulation; and examined the triggers and deleterious immuno-inflammatory consequences. In the present study, we found that circulating NET-related biomarker levels increased in serum and blister fluid of BP patients and significantly correlated with disease severity. Additionally, circulating neutrophils from BP patients displayed enhanced spontaneous NETs formation than healthy controls. In vitro, BP180-NC16A immune complexes-induced NETosis in neutrophils from BP patients, which was abrogated by Fcγ receptor and/or NADPH pathway blockade. Furthermore, the elevated levels of NETs from BP patients boosted autoantibody production by inducing B-cell differentiation into plasma cells, mediated by MAPK P38 cascade activation. Together, our findings provide strong evidence that NETs are involved in a pathogenic loop, causing excessive differentiation of B cells and promotion of autoantibody production. Hence, targeting aberrant neutrophil responses will provide novel potential targets for the treatment of BP.
Collapse
Affiliation(s)
- Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke Xue
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xu Yuan
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jieyu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tianyu Cao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yixin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaocui Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjing Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Caixia Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongjiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
42
|
|
43
|
Cui M, Li T, Yan X, Wang C, Shen Q, Ren H, Li L, Zhang R. Blood Genomics Identifies Three Subtypes of Systemic Lupus Erythematosus: "IFN-High," "NE-High," and "Mixed". Mediators Inflamm 2021; 2021:6660164. [PMID: 34305454 PMCID: PMC8270691 DOI: 10.1155/2021/6660164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/29/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Systemic lupus erythematosus (SLE) is a systemic and multifactorial autoimmune disease, and its diverse clinical manifestations affect molecular diagnosis and drug benefits. Our study was aimed at defining the SLE subtypes based on blood transcriptome data, analyzing functional patterns, and elucidating drug benefits. METHODS Three data sets were used in this paper that were collected from the Gene Expression Omnibus (GEO) database, which contained two published data sets of pediatric and adult SLE patients (GSE65391, GSE49454) and public longitudinal data (GSE72754) from a cohort of SLE patients treated with IFN-α Kinoid (IFN-K). Based on disease activity scores and gene expression data, we defined a global SLE signature and merged three clustering algorithms to develop a single-sample subtype classifier (SSC). Systematic analysis of coexpression networks based on modules revealed the molecular mechanism for each subtype. RESULTS We identified 92 genes as a signature of the SLE subtypes and three intrinsic subsets ("IFN-high," "NE-high," and "mixed"), which varied in disease severity. We speculated that IFN-high might be due to the overproduction of interferons (IFNs) caused by viral infection, leading to the formation of autoantibodies. NE-high might primarily result from bacterial and fungal infections that stimulated neutrophils (NE) to produce neutrophil extracellular traps (NETs) and induced individual autoimmune responses. The mixed type contained both of these molecular mechanisms and showed an intrinsic connection. CONCLUSIONS Our research results indicated that identifying the molecular mechanism associated with different SLE subtypes would benefit the molecular diagnosis and stratified therapy. Moreover, repositioning of IFN-K based on subtypes also revealed an improved therapeutic effect, providing a new direction for disease treatment and drug development.
Collapse
Affiliation(s)
- Mintian Cui
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Taotao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Xinwei Yan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Chao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Qi Shen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Hongbiao Ren
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Liangshuang Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| |
Collapse
|
44
|
Müller A, Krause B, Kerstein-Stähle A, Comdühr S, Klapa S, Ullrich S, Holl-Ulrich K, Lamprecht P. Granulomatous Inflammation in ANCA-Associated Vasculitis. Int J Mol Sci 2021; 22:ijms22126474. [PMID: 34204207 PMCID: PMC8234846 DOI: 10.3390/ijms22126474] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022] Open
Abstract
ANCA-associated vasculitis (AAV) comprises granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). While systemic vasculitis is a hallmark of all AAV, GPA is characterized by extravascular granulomatous inflammation, preferentially affecting the respiratory tract. The mechanisms underlying the emergence of neutrophilic microabscesses; the appearance of multinucleated giant cells; and subsequent granuloma formation, finally leading to scarred or destroyed tissue in GPA, are still incompletely understood. This review summarizes findings describing the presence and function of molecules and cells contributing to granulomatous inflammation in the respiratory tract and to renal inflammation observed in GPA. In addition, factors affecting or promoting the development of granulomatous inflammation such as microbial infections, the nasal microbiome, and the release of damage-associated molecular patterns (DAMP) are discussed. Further, on the basis of numerous results, we argue that, in situ, various ways of exposure linked with a high number of infiltrating proteinase 3 (PR3)- and myeloperoxidase (MPO)-expressing leukocytes lower the threshold for the presentation of an altered PR3 and possibly also of MPO, provoking the local development of ANCA autoimmune responses, aided by the formation of ectopic lymphoid structures. Although extravascular granulomatous inflammation is unique to GPA, similar molecular and cellular patterns can be found in both the respiratory tract and kidney tissue of GPA and MPA patients; for example, the antimicrobial peptide LL37, CD163+ macrophages, or regulatory T cells. Therefore, we postulate that granulomatous inflammation in GPA or PR3-AAV is intertwined with autoimmune and destructive mechanisms also seen at other sites.
Collapse
Affiliation(s)
- Antje Müller
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Correspondence: ; Tel.: +49-451-5005-0867
| | - Bettina Krause
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Institute of Anatomy & Experimental Morphology, University Hospital Hamburg-Eppendorf, University of Hamburg, 20251 Hamburg, Germany;
| | - Anja Kerstein-Stähle
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| | - Sara Comdühr
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| | - Sebastian Klapa
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
- Institute of Experimental Medicine c/o German Naval Medical Institute, Carl-Albrechts University of Kiel, 24119 Kronshagen, Germany
| | - Sebastian Ullrich
- Institute of Anatomy & Experimental Morphology, University Hospital Hamburg-Eppendorf, University of Hamburg, 20251 Hamburg, Germany;
- Municipal Hospital Kiel, 24116 Kiel, Germany
| | | | - Peter Lamprecht
- Department of Rheumatology & Clinical Immunology, University of Luebeck, 23562 Luebeck, Germany; (B.K.); (A.K.-S.); (S.C.); (S.K.); (P.L.)
| |
Collapse
|
45
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
46
|
Physical exercise, obesity, inflammation and neutrophil extracellular traps (NETs): a review with bioinformatics analysis. Mol Biol Rep 2021; 48:4625-4635. [PMID: 34014471 DOI: 10.1007/s11033-021-06400-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/04/2021] [Indexed: 12/23/2022]
Abstract
Neutrophil extracellular traps (NETs) represent an innate organism defense mechanism characterized by neutrophil release of intracellular material to capture any aggressor agent. Elevated NETs release is associated with increased inflammatory response and related diseases, such as obesity. Chronic physical training is one of the main strategies to treat and prevent obesity. The relationship between physical training and NETs is still under study. The present review, followed by a bioinformatics analysis, demonstrates the meaningful connection between physical exercise, obesity, and NETs. The bioinformatics indicated TNF-α as a leading gene after the ontological analysis followed by positive-interleukin-6 regulation, chemokines, and inflammatory response regulation. The main results pointed to a relevant regulatory effect of physical training on NETs release, indicating physical exercise as a possible therapeutic target on modulating NETs and inflammation.
Collapse
|
47
|
Li J, Wang C, Liu J, Yu Y, Liu Y, Peng Q, Liu H, Guan X. A feedback loop: Interactions between Inflammatory Signals and Clonal Hematopoiesis in Cardiovascular Disease. Mol Biol Rep 2021; 48:3785-3798. [PMID: 33987748 PMCID: PMC8117808 DOI: 10.1007/s11033-021-06370-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
Age and inflammation are powerful drivers of cardiovascular disease. With the growing recognition that traditional cardiovascular risk factors are not fully accurate predictors of cardiovascular disease, recent studies have revealed the prevalence of positive selection of somatic cell mutations in hematopoietic stem cells in the elderly population, which can cause clonal hematopoiesis. Interestingly, clonal hematopoiesis is not only associated with cancer and death, but also closely related to the risk of increased cardiovascular disease due to mutations in TET2, DNMT3A, ASXL1, and JAK2. However, the mechanism of the interaction of clonal hematopoiesis and cardiovascular disease is only partially understood. In mice, somatic mutations have led to significantly increased expression of inflammatory genes in innate immune cells, which may explain the relationship between mutations and cardiovascular disease. Here, we further discuss the association between inflammatory signaling, clonal hematopoiesis, and cardiovascular disease,and using two hypotheses to propose a feedback loop between inflammatory signaling and clonal hematopoiesis for getting insight into the pathogenesis of cardiovascular diseases in depth. Therapies targeting mutant clones or increased inflammatory mediators may be useful for ameliorating the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Jiashan Li
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Chao Wang
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jiaru Liu
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ying Yu
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yuee Liu
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Qi Peng
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Huihui Liu
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiuru Guan
- First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
48
|
Bautista-Becerril B, Campi-Caballero R, Sevilla-Fuentes S, Hernández-Regino LM, Hanono A, Flores-Bustamante A, González-Flores J, García-Ávila CA, Aquino-Gálvez A, Castillejos-López M, Juárez-Cisneros A, Camarena A. Immunothrombosis in COVID-19: Implications of Neutrophil Extracellular Traps. Biomolecules 2021; 11:694. [PMID: 34066385 PMCID: PMC8148218 DOI: 10.3390/biom11050694] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is a member of the family of coronaviruses associated with severe outbreaks of respiratory diseases in recent decades and is the causative agent of the COVID-19 pandemic. The recognition by and activation of the innate immune response recruits neutrophils, which, through their different mechanisms of action, form extracellular neutrophil traps, playing a role in infection control and trapping viral, bacterial, and fungal etiological agents. However, in patients with COVID-19, activation at the vascular level, combined with other cells and inflammatory mediators, leads to thrombotic events and disseminated intravascular coagulation, thus leading to a series of clinical manifestations in cerebrovascular, cardiac, pulmonary, and kidney disease while promoting severe disease and mortality. Previous studies of hospitalized patients with COVID-19 have shown that elevated levels of markers specific for NETs, such as free DNA, MPO, and H3Cit, are strongly associated with the total neutrophil count; with acute phase reactants that include CRP, D-dimer, lactate dehydrogenase, and interleukin secretion; and with an increased risk of severe COVID-19. This study analyzed the interactions between NETs and the activation pathways involved in immunothrombotic processes in patients with COVID-19.
Collapse
Affiliation(s)
- Brandon Bautista-Becerril
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
- Programa MEDICI, Carrera Médico Cirujano, FES Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (R.C.-C.); (J.G.-F.)
| | - Rebeca Campi-Caballero
- Programa MEDICI, Carrera Médico Cirujano, FES Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (R.C.-C.); (J.G.-F.)
- Laboratorio de Neuropsicofarmacología, Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Samuel Sevilla-Fuentes
- Departamento de Infectología, Hospital General de México Eduardo Liceaga, Mexico City 06720, Mexico;
| | - Laura M. Hernández-Regino
- Escuela Nacional de Ciencias Biológicas, Programa de Posgrado, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (L.M.H.-R.); (C.A.G.-Á.)
| | - Alejandro Hanono
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Mexico City 52786, Mexico;
| | - Al Flores-Bustamante
- Laboratorio de Farmacología, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Julieta González-Flores
- Programa MEDICI, Carrera Médico Cirujano, FES Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (R.C.-C.); (J.G.-F.)
| | - Carlos A. García-Ávila
- Escuela Nacional de Ciencias Biológicas, Programa de Posgrado, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (L.M.H.-R.); (C.A.G.-Á.)
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Manuel Castillejos-López
- Departamento de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Armida Juárez-Cisneros
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
| | - Angel Camarena
- Laboratorio HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (B.B.-B.); (A.J.-C.)
| |
Collapse
|
49
|
Barbu EA, Dominical VM, Mendelsohn L, Thein SL. An Imaging Flow Cytometry Method to Measure Citrullination of H4 Histone as a Read-out for Neutrophil Extracellular Traps Formation. Bio Protoc 2021; 11:e3927. [PMID: 33732814 DOI: 10.21769/bioprotoc.3927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/20/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
The formation of neutrophil extracellular traps (NETs) is thought to play a critical role in infections and propagating sterile inflammation. Histone citrullination is an essential and early step in NETs formation, detectable prior to the formation of the hallmark extracellular DNA-scaffolded strands. In addition to the classical microscopy method, new technologies are being developed for studies of NETs and their detection, both for research and clinical purposes. Classical microscopy studies of NETs are subjective, low throughput and semi-quantitative, and limited in their ability to capture the early steps. We have developed this novel Imaging Flow Cytometry (IFC) method that specifically identifies and quantifies citrullination of histone H4 as a NETs marker and its relationship with other alterations at nuclear and cellular level. These include nuclear decondensation and super-condensation, multi-lobulated nuclei versus 1-lobe nuclei and cell membrane damage. NETs markers can be quantified following variable periods of treatment with NETs inducers, prior to the formation of the specific extracellular DNA-scaffolded strands. Because these high throughput image-based cell analysis features can be performed with statistical rigor, this protocol is suited for both experimental and clinical applications as well as clinical evaluations of NETosis as a biomarker.
Collapse
Affiliation(s)
- Emilia A Barbu
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, USA
| | - Venina M Dominical
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, NIH, Bethesda, USA
| | - Laurel Mendelsohn
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, USA
| |
Collapse
|
50
|
Blindar VN, Zubrikhina GN, Sytov AV. Morphological and functional features of peripheral blood neutrophils in cancer patients with sepsis in the early postoperative period (review of literature and results of our own studies). Klin Lab Diagn 2021; 66:15-21. [PMID: 33567168 DOI: 10.18821/0869-2084-2021-66-1-15-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The study was performed in 9 cancer patients with sepsis. A clinical blood test of patients with sepsis was accompanied by a neutrophilic leukemoid reaction, with the appearance in the differential count of white blood cells of young cells (myelocytes, metamyelocytes). According to the literature, it is shown that in severe sepsis, a local infection is accompanied by systemic of neutrophil activation (NF). Moreover, excessive activation and location of NF in the microvasculature, especially its young forms and the formation of extracellular trap networks (NETs), can contribute to pathological manifestations of multiple organ failure and thrombosis.
Collapse
Affiliation(s)
| | - G N Zubrikhina
- FSBI «National Medical Research Center of Oncology N.N. Blokhin» Ministry of Health of Russia
| | - A V Sytov
- FSBI «National Medical Research Center of Oncology N.N. Blokhin» Ministry of Health of Russia
| |
Collapse
|