1
|
Zhou M, Lu Y, Tang Y, Zhang T, Xiao D, Zhang M, Zhang S, Li J, Cai X, Lin Y. A DNA-based nanorobot for targeting, hitchhiking, and regulating neutrophils to enhance sepsis therapy. Biomaterials 2025; 318:123183. [PMID: 39951831 DOI: 10.1016/j.biomaterials.2025.123183] [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: 11/20/2024] [Revised: 12/29/2024] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
Targeted regulation of neutrophils is an effective approach for treating neutrophil-driven inflammatory diseases, but challenges remain in minimizing off-target effects and extending drug half-life. A DNA-based nanorobot was developed to target neutrophils by using an N-acetyl Pro-Gly-Pro (Ac-PGP) peptide to specifically bind to the C-X-C motif of chemokine receptor 2 (CXCR2) on neutrophil membranes. This robot (a tetrahedral framework nucleic acid modified with Ac-PGP, APT) identified and hitchhiked neutrophils to accumulate at inflammatory sites and prolong its half-lives, whilst also was internalized to influence the neutrophil cell cycle and maturation process to regulate oxidative stress, inflammation, migration, and recruitment in both in vivo and in vitro inflammation experiments. Consequently, the tissue damage caused by sepsis was greatly reduced. This novel neutrophil-based nanorobot highlights the high precision of targeting and regulating neutrophils, and presents a potential strategy for treating multiple neutrophil-driven diseases.
Collapse
Affiliation(s)
- Mi Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanlin Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shunhao Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China; National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
2
|
Till JE, Seewald NJ, Yazdani Z, Wang Z, Ballinger D, Samberg H, Dandu S, Macia C, Yin M, Abdalla A, Prior T, Shah SS, Patel T, McCoy E, Mansour M, Wills CA, Bochenek V, Serrano J, Snuderl M, Phillips RE, O'Rourke DM, Amankulor NM, Nabavizadeh A, Desai AS, Gollomp K, Binder ZA, Zhou W, Bagley SJ, Carpenter EL. Corticosteroid-Dependent Association between Prognostic Peripheral Blood Cell-Free DNA Levels and Neutrophil-Mediated NETosis in Patients with Glioblastoma. Clin Cancer Res 2025; 31:1292-1304. [PMID: 39887264 DOI: 10.1158/1078-0432.ccr-24-3169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/13/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
PURPOSE Noninvasive prognostic biomarkers to inform clinical decision-making are an urgent unmet need for the management of patients with glioblastoma (GBM). We previously showed that higher circulating cell-free DNA (ccfDNA) concentration is associated with worse survival in GBM. However, the biology underlying this is unknown. EXPERIMENTAL DESIGN We prospectively enrolled 129 patients with treatment-naïve GBM with blood drawn prior to initial resection (baseline) and at the time of the first postradiotherapy MRI. We performed ccfDNA methylation deconvolution to determine cellular sources of ccfDNA. ELISA was performed to detect citrullinated histone 3 (citH3), a marker of neutrophil extracellular traps (NET). Multiplex proteomic analysis was used to measure soluble inflammatory proteins. RESULTS We found that neutrophils contributed the highest proportion of prognostic ccfDNA. The percentage of ccfDNA derived from neutrophils was correlated with total [ccfDNA] but only in patients receiving preoperative corticosteroids. At baseline and on therapy, [citH3] was significantly higher in the plasma of patients with GBM receiving corticosteroids compared with corticosteroid-naïve GBM or no-cancer controls. Unsupervised hierarchical clustering of ccfDNA methylation patterns yielded two clusters, with one enriched for patients with the NETosis phenotype and who received corticosteroids. Unsupervised clustering of circulating inflammatory proteins yielded similar results. CONCLUSIONS These data suggest neutrophil-mediated NETosis is the dominant source of prognostic ccfDNA in patients with GBM and may be associated with glucocorticoid exposure. If further studies show that pharmacological inhibition of NETosis can mitigate the deleterious effects of corticosteroids, these plasma markers will have important clinical utility as noninvasive correlative biomarkers.
Collapse
Affiliation(s)
- Jacob E Till
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nicholas J Seewald
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zachariya Yazdani
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zhuoyang Wang
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Dominique Ballinger
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Heather Samberg
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Siri Dandu
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Camilla Macia
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Melinda Yin
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Aseel Abdalla
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Timothy Prior
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shivani S Shah
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Thara Patel
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Emily McCoy
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Maikel Mansour
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Carson A Wills
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Veronica Bochenek
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jonathan Serrano
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Matija Snuderl
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Richard E Phillips
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Donald M O'Rourke
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nduka M Amankulor
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ali Nabavizadeh
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Arati S Desai
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kandace Gollomp
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Zev A Binder
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Wanding Zhou
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Stephen J Bagley
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Erica L Carpenter
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Zhou E, Li Y, Wu Z, Chen Y, Wu H, Ye Y, Li T, Wang J, Yang Z. Neutrophil extracellular traps formation and autophagy in bongkrekic acid exposed human neutrophils. Toxicol In Vitro 2025; 104:106003. [PMID: 39730015 DOI: 10.1016/j.tiv.2024.106003] [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: 01/03/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024]
Abstract
Bongkrekic acid (BKA), a less well-known foodborne toxin, has been implicated in numerous poisoning incidents. Recent studies suggest that BKA exerts an impact on the immune system, particularly on innate immunity. The release of neutrophil extracellular traps (NETs) is relatively a newly-discovered mechanism involving innate immunity. This study was designed to characterize and evaluate the effects of BKA on human NET formation. The co-localization of DNA, histones, and myeloperoxidase (MPO) was determined via immunostaining to confirm BKA-triggered NET formation in human neutrophils. NET quantification showed that NET formation induced by BKA was both time- and dose-dependent, and was associated with p38, ERK, PAD4 and P2X1 receptor. Moreover, immunostaining analysis observed that BKA triggered both NET formation and autophagy. Additionally, pharmacological experiments revealed that autophagy mediated BKA-triggered NET formation. Collectively, these insights offer a novel perspective on the effects of BKA exposure on host's innate immune response, and may shed new light on BKA poisoning. We call for further work to be conducted in this field to unravel the intricate mechanisms governing NET formation and autophagy in the context of BKA poisoning.
Collapse
Affiliation(s)
- Ershun Zhou
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yifei Li
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhikai Wu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yichun Chen
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Hanpeng Wu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yingrong Ye
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Tianyu Li
- Guangxi University, Nanning 530004, Guangxi Province, PR China
| | - Jingjing Wang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China.
| | - Zhengtao Yang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China.
| |
Collapse
|
4
|
Varjú I, Tanka-Salamon A, Kolev K. Neutrophil Extracellular Traps: At the Interface of Thrombosis and Comorbidities. Semin Thromb Hemost 2025. [PMID: 40020757 DOI: 10.1055/a-2548-0805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Since their discovery in 2004, neutrophil extracellular traps (NETs) have been at the center of multidisciplinary attention. Although a key tool in neutrophil-mediated immunity, these filamentous, enzyme-enriched DNA-histone complexes can be detrimental to tissues and have been identified as an underlying factor in a range of pathological conditions. Building on more than 20 years of research into NETs, this review places thrombosis, the pathological formation of blood clots, in the spotlight. From this point of view, we discuss the structure and formation of NETs, as well as the interaction of their components with the hemostatic system, dissecting the pathways through which NETs exert their marked effect on formation and the dissolution of thrombi. We pay distinct attention to the latest developments in the research of a key player in NET formation, peptidyl-arginine-deiminase (PAD) enzymes: their types, sources, and potential cross-play with the hemostatic machinery. Besides these molecular details, we elaborate on the link between pathological thrombosis, NETs, and widespread conditions that represent a debilitating public health burden worldwide, such as sepsis and neoplasms. Finally, future implications on the treatment of thrombosis-related conditions will be discussed.
Collapse
Affiliation(s)
- Imre Varjú
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Anna Tanka-Salamon
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Zhang P, Han J, Kong X, Liu S, Chen Y, Li J, Zhang Y, Wang C, Du L. Biomimetic Synthesis of Nanomachine Inspired from Neutrophil Extracellular Traps for Multimodal Antibacterial Application. ACS NANO 2025. [PMID: 40134237 DOI: 10.1021/acsnano.4c18948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Bacterial infections, especially drug-resistant bacterial infections, are causing increasing harm in clinical practice, and there is an urgent need to develop effective antimicrobial materials. Biomimetic DNA nanomachines have attracted much attention due to their flexible design, precise control, and high biocompatibility, but their use for bacterial inhibition has not been reported. Neutrophil extracellular traps (NETs), a network structure released by neutrophils with good bactericidal function, can be used as a superior biomimetic object for the construction of functional bacterial inhibition materials. In this study, Y-shaped DNA was polymerized using magnesium ions to develop reticulated DNA structures, which were used as templates to synthesize copper nanoclusters, leading to the construction of compositionally well-defined and simple reticulated DNA nanomachines. The nanomachine had a three-dimensional, reticular structure similar to that of NETs and especially had excellent antibacterial activity. More importantly, the NETs-imitated nanomachine had a multimodal bacterial inhibition mechanism. The nanomachine could target and localize around the bacteria and eliminate the biofilm, and then the DNA network structure effectively trapped and aggregated the bacteria and caused damage to the bacterial morphology and membrane structure; at the same time, the reticulated DNA nanomachine could also damage the bacterial membrane, causing the degradation and leakage of the proteins and the cellular contents and breakage of the DNA structure, ultimately causing irreversible inhibition of the bacteria. Importantly, the developed nanomachines with high biocompatibility could be used as antimicrobial biomaterials for the efficient treatment and healing of skin wounds infected with bacteria. This study develops a biomimetic DNA nanomachine that can be an excellent antibacterial biomaterial, which expands the application of DNA nanomachine in bacteriostatic and therapeutic fields; it is also an improved biomimetic NETs biomaterial, which brings distinctive design sources for biomimetic materials.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Jinxiu Han
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
- Department of Clinical Laboratory, Children's Hospital Affiliated to Shandong University, Jinan 250022, China
| | - Xue Kong
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Shaojun Liu
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Yuqing Chen
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Juan Li
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
| | - Lutao Du
- Department of Clinical Laboratory, Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker Detection the Second Hospital of Shandong University, Jinan 250033, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan 250012, China
| |
Collapse
|
6
|
Wu S, Chen Y, Jin X, Yu J, Chen X, Wan T. Toll Like Receptors Promote High Glucose-Induced Vascular Endothelial Cell Dysfunction by Regulating Neutrophil Extracellular Traps Formation. Inflammation 2025:10.1007/s10753-025-02283-8. [PMID: 40087251 DOI: 10.1007/s10753-025-02283-8] [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: 09/07/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Diabetic retinopathy (DR) is a major cause of blindness globally. Neutrophils and neutrophil extracellular traps (NETs) are believed to play a role in the development of DR. However, the specific contribution of NETs to hyperglycemia-induced vascular endothelial cell dysfunction remains unclear. In this study, we cocultured high glucose-activated neutrophils (HGNs) with human umbilical vein endothelial cells (HUVECs) to investigate the role of NETs in high glucose-induced HUVEC dysfunction. Our findings indicate that high glucose levels promote NETs formation, which can be inhibited by a toll-like receptor (TLR) 2 antagonist and a TLR4 antagonist. It was observed that reactive oxygen species production plays a role in TLR2- but not TLR4-mediated NETs formation. Additionally, HGNs were found to promote HUVEC proliferation through phagocytosis rather than NETs. We also discovered that NETs contribute to high glucose-induced HUVEC dysfunction by enhancing neutrophil-HUVEC adhesion, inhibiting HUVEC migration, and compromising the barrier function of the cells by reducing zonula occludens-1 expression. This dysfunction could be partially mitigated by TLR2 and TLR4 antagonists. In conclusion, high glucose stimulates NETs formation, leading to vascular endothelial cell damage, and TLRs may facilitate high glucose-induced endothelial dysfunction by modulating NETs formation.
Collapse
Affiliation(s)
- Shirou Wu
- Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Yahui Chen
- Quintiles Medical Research & Development Beijing Ltd, Liangshuihe No.2 Street, Beijing, 100176, China
| | - Xiuming Jin
- Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Jiayun Yu
- Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Xueping Chen
- Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China
| | - Ting Wan
- Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| |
Collapse
|
7
|
Larsen ML, Nørgaard L, Linge P, Larsen JB, Langkilde HZ, Hauge EM, Thiel S, Voss A, Bengtsson A, Troldborg A. Molecular mechanisms underlying thrombosis in systemic lupus erythematosus - A Systematic review. Semin Arthritis Rheum 2025; 72:152707. [PMID: 40086157 DOI: 10.1016/j.semarthrit.2025.152707] [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: 10/03/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
Patients with systemic lupus erythematosus (SLE) face an approximately 30 % risk of thrombosis post-diagnosis. However, there remains significant knowledge gaps regarding causative mechanisms, and there is a lack of specific antithrombotic guidelines. This systematic review aims to examine the existing literature regarding the mechanisms contributing to thrombosis risk in SLE, focusing on five predefined procoagulant domains: autoantibodies (including antiphospholipid antibodies (aPL)), the complement system, platelets, the endothelium, and the coagulation system. The review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statements and searched in PubMed and Embase without time restrictions. Risk of bias assessment was performed using a pre-specified evaluation tool. Out of 3,747 initially identified publications, 30 studies were included, with 28 demonstrating robust methodological quality in the risk of bias assessment. The studies were experimental, involving blood samples from cross-sectional SLE cohorts, except one animal -and one case-control study. We identified six different thrombosis mechanisms of action. Most studies concentrated on autoantibodies, predominantly aPL. Shared mechanisms between aPL and other autoantibodies may account for the increased thrombosis risk in aPL-negative SLE patients. Significant knowledge gaps remain, particularly regarding the role of the complement system in SLE-related thrombosis. Also, most research relies on cross-sectional designs, emphasizing the need for prospective cohort studies to better assess clinical factors. Finally, comprehensive studies examining the interactions between multiple procoagulant factors and their link to thrombosis are lacking. Closing these gaps in future research could improve both preventive and personalized treatment strategies for thrombosis in SLE.
Collapse
Affiliation(s)
- Mads L Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Laura Nørgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Petrus Linge
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Julie B Larsen
- Department of Clinical Biochemistry, Regional Hospital Horsens, Horsens, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henrik Z Langkilde
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Ellen M Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anne Voss
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Anders Bengtsson
- Department of Clinical Sciences, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anne Troldborg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
8
|
Shi Z, Gong S, Li Y, Yan K, Bao Y, Ning K. Neutrophil Extracellular Traps in Atherosclerosis: Research Progress. Int J Mol Sci 2025; 26:2336. [PMID: 40076955 PMCID: PMC11900999 DOI: 10.3390/ijms26052336] [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: 01/13/2025] [Revised: 02/15/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Atherosclerosis (AS) is a disease characterised by the accumulation of atherosclerotic plaques on the inner walls of blood vessels, resulting in their narrowing. In its early stages, atherosclerosis remains asymptomatic and undetectable by conventional pathological methods. However, as the disease progresses, it can lead to a series of cardiovascular diseases, which are a leading cause of mortality among middle-aged and elderly populations worldwide. Neutrophil extracellular traps (NETs) are composed of chromatin and granular proteins released by neutrophils. Upon activation by external stimuli, neutrophils undergo a series of reactions, resulting in the release of NETs and subsequent cell death, a process termed NETosis. Research has demonstrated that NETosis is a means by which neutrophils contribute to immune responses. However, studies on neutrophil extracellular traps have identified NETs as the primary cause of various inflammation-induced diseases, including cystic fibrosis, systemic lupus erythematosus, and rheumatoid arthritis. Consequently, the present review will concentrate on the impact of neutrophil extracellular traps on atherosclerosis formation, analysing it from a molecular biology perspective. This will involve a systematic dissection of their proteomic components and signal pathways.
Collapse
Affiliation(s)
- Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (Z.S.); (S.G.); (Y.L.); (K.Y.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, Preston Research Building, Room 359, Nashville, TN 37232, USA
| |
Collapse
|
9
|
Zalghout S, Martinod K. Therapeutic potential of DNases in immunothrombosis: promising succor or uncertain future? J Thromb Haemost 2025; 23:760-778. [PMID: 39667687 DOI: 10.1016/j.jtha.2024.11.028] [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: 09/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
Sepsis, a life-threatening condition characterized by systemic inflammation and multiorgan dysfunction, is closely associated with the excessive formation of neutrophil extracellular traps (NETs) and the release of cell-free DNA. Both play a central role in sepsis progression, acting as major contributors to immunothrombosis and associated complications. Endogenous DNases play a pivotal role in degrading NETs and cell-free DNA, yet their activity is often dysregulated during thrombotic disease. Although exogenous DNase1 administration has shown potential in reducing NET burden and mitigating the detrimental effects of immunothrombosis, its therapeutic efficacy upon intravenous administration remains uncertain. The development of engineered DNase formulations and combination therapies may further enhance its therapeutic effectiveness by modifying its pharmacodynamic properties and avoiding the adverse effects associated with NET degradation, respectively. Although NETs are well-established targets of DNase1, it remains uncertain whether the positive effects of DNase1 on immunothrombosis are exclusively related to it's targeting of NETs or if other components contributing to immunothrombosis are also affected. This review examines the endogenous regulation of NETs in circulation and the therapeutic potential of DNases in immunothrombosis, underscoring the necessity for further investigation to optimize their clinical application.
Collapse
Affiliation(s)
- Sara Zalghout
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
10
|
Potempa M, Hart PC, Rajab IM, Potempa LA. Redefining CRP in tissue injury and repair: more than an acute pro-inflammatory mediator. Front Immunol 2025; 16:1564607. [PMID: 40093010 PMCID: PMC11906453 DOI: 10.3389/fimmu.2025.1564607] [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: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Most early studies investigating the role of C-reactive protein (CRP) in tissue damage determined it supported pro-hemostatic and pro-inflammatory activities. However, these findings were not universal, as other data suggested CRP inhibited these same processes. A potential explanation for these disparate observations finally emerged with the recognition that CRP undergoes context-dependent conformational changes in vivo, and each of its three isoforms - pentameric CRP (pCRP), modified pentameric CRP (pCRP*), and monomeric CRP (mCRP) - have different effects. In this review, we consider this new paradigm and re-evaluate the role of CRP and its isoforms in the tissue repair process. Indeed, a growing body of evidence points toward the involvement of CRP not just in hemostasis and inflammation, but also in the resolution of inflammation and in tissue regeneration. Additionally, we briefly discuss the shortcomings of the currently available diagnostic tests for CRP and highlight the need for change in how CRP is currently utilized in clinical practice.
Collapse
Affiliation(s)
| | - Peter C. Hart
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| | - Ibraheem M. Rajab
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| | - Lawrence A. Potempa
- Acphazin Inc., Deerfield, IL, United States
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| |
Collapse
|
11
|
Li H, Shan W, Zhao X, Sun W. Neutrophils: Linking Inflammation to Thrombosis and Unlocking New Treatment Horizons. Int J Mol Sci 2025; 26:1965. [PMID: 40076593 PMCID: PMC11901051 DOI: 10.3390/ijms26051965] [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: 01/02/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophils play a key role in inflammatory responses and thrombosis, but their complex interactions in disease pathogenesis are not fully understood. This review examines the multifaceted roles of neutrophils, focusing on their activation, cytokine release, and formation of neutrophil extracellular traps (NETs), which contribute to host defense and thrombosis. We discuss the interaction between inflammation and coagulation, the direct effect of neutrophils on thrombus stability, and their involvement in pathological thrombotic diseases. The therapeutic potential of neutrophil drug loading in the treatment of thrombosis, as well as the clinical implications and future research directions, are highlighted. The aim of this review is to gain insight into the critical neutrophil-inflammation-thrombus axis and its potential as a therapeutic target for thrombotic diseases and to suggest possible directions for neutrophil-loaded drug therapy for thrombosis.
Collapse
Affiliation(s)
| | | | | | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.L.); (W.S.); (X.Z.)
| |
Collapse
|
12
|
Sennett C, Pula G. Trapped in the NETs: Multiple Roles of Platelets in the Vascular Complications Associated with Neutrophil Extracellular Traps. Cells 2025; 14:335. [PMID: 40072064 PMCID: PMC11898727 DOI: 10.3390/cells14050335] [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: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Neutrophil extracellular traps (NETs) have received significant attention in recent years for their role in both the immune response and the vascular damage associated with inflammation. Platelets have been described as critical components of NETs since the initial description of this physio-pathological response of neutrophils. Platelets have been shown to play a dual role as responders and also as stimulators of NETs. The direct interaction with DNA leads to the entrapment of platelets into NETs, a phenomenon that significantly contributes to the thrombotic complications of inflammation and neutrophil activation, while the direct and paracrine stimulation of neutrophils by platelets has been shown to initiate the process of NET formation. In this review, we provide a comprehensive description of our current understanding of the molecular mechanisms underlying the entrapping of platelets into NETs and, in parallel, the platelet-driven cellular responses promoting NET formation. We then illustrate established examples of the contribution of NETs to vascular pathologies, describe the important questions that remain to be answered regarding the contribution of platelets to NET formation and NET-dependent cardiovascular complication, and highlight the fundamental steps taken towards the application of our understanding of platelets' contribution to NETs for the development of novel cardiovascular therapies.
Collapse
Affiliation(s)
| | - Giordano Pula
- Biomedical Institute for Multimorbidity (BIM), Hull York Medical School (HYMS), University of Hull, Hull HU6 7RX, UK
| |
Collapse
|
13
|
Pène F, Russell L, Aubron C. Thrombocytopenia in the intensive care unit: diagnosis and management. Ann Intensive Care 2025; 15:25. [PMID: 39985745 PMCID: PMC11846794 DOI: 10.1186/s13613-025-01447-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/09/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND This narrative review aims to describe the epidemiology and aetiologies of thrombocytopenia in critically ill patients, the bleeding risk assessment in thrombocytopenic patients, and provide an update on platelet transfusion indications. RESULTS Thrombocytopenia is a common disorder in critically ill patients. The classic definition relies on an absolute platelet count below 150 × 109/L. Alternatively, the definition has extended to a relative decrease in platelet count (typically within a range of >30->50% decrease) from baseline, yet remaining above 150 × 109/L. Thrombocytopenia may result from multiple mechanisms depending upon the underlying conditions and the current clinical setting. Regardless of the causes, thrombocytopenia accounts as an independent determinant of poor outcomes in critically ill patients, albeit often of unclear interpretation. Nevertheless, it is well established that thrombocytopenia is associated with an increased incidence of bleeding complications. However, alternative factors also contribute to the risk of bleeding, making it difficult to establish definite links between nadir platelet counts at the expense of potential adverse events. Platelet transfusion represents the primary supportive treatment of thrombocytopenia to prevent or treat bleeding. As randomised controlled trials comparing different platelet count thresholds for prophylactic platelet transfusion in the ICU are lacking, the prophylactic transfusion strategy is largely derived from studies performed in stable haematology patients. Similarly, the platelet count transfusion threshold to secure invasive procedures remains based on a low level of evidence. Indications of platelet transfusions for the treatment of severe bleeding in thrombocytopenic patients remain largely empirical, with platelet count thresholds ranging from 50 to 100 × 109/L. In addition, early and aggressive platelet transfusion is part of massive transfusion protocols in the setting of severe trauma-related haemorrhage. CONCLUSION Thrombocytopenia in critically ill patients is very frequent with various etiologies, and is associated with worsened prognosis, with or without bleeding complications. Interventional trials focused on critically ill patients are eagerly needed to better delineate the benefits and harms of platelet transfusions.
Collapse
Affiliation(s)
- Frédéric Pène
- Service de Médecine Intensive - Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre, Université Paris Cité, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France.
| | - Lene Russell
- Department of Intensive Care, Copenhagen University Hospital Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Cécile Aubron
- Service de Médecine Intensive - Réanimation, CHU de Brest, Université de Bretagne Occidentale, Brest, France
| |
Collapse
|
14
|
Bryzek D, Gasiorek A, Kowalczyk D, Santocki M, Ciaston I, Dobosz E, Kolaczkowska E, Kjøge K, Kantyka T, Lech M, Potempa B, Enghild JJ, Potempa J, Koziel J. Non-classical neutrophil extracellular traps induced by PAR2-signaling proteases. Cell Death Dis 2025; 16:109. [PMID: 39971938 PMCID: PMC11840154 DOI: 10.1038/s41419-025-07428-z] [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: 09/09/2024] [Revised: 01/21/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Neutrophil extracellular traps (NETs) are associated with diseases linked to aberrant coagulation. The blood clotting cascade involves a series of proteases, some of which induce NET formation via a yet unknown mechanism. We hypothesized that this formation involves signaling via a factor Xa (FXa) activation of the protease-activated receptor 2 (PAR2). Our findings revealed that NETs can be triggered in vitro by enzymatically active proteases and PAR2 agonists. Intravital microscopy of the liver vasculature revealed that both FXa infusion and activation of endogenous FX promoted NET formation, effects that were prevented by the FXa inhibitor, apixaban. Unlike classical NETs, these protease-induced NETs lacked bactericidal activity and their proteomic signature indicates their role in inflammatory disorders, including autoimmune diseases and carcinogenesis. Our findings suggest a novel mechanism of NET formation under aseptic conditions, potentially contributing to a self-amplifying clotting and NET formation cycle. This mechanism may underlie the pathogenesis of disseminated intravascular coagulation and other aseptic conditions.
Collapse
Affiliation(s)
- Danuta Bryzek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Anna Gasiorek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dominik Kowalczyk
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michal Santocki
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Izabela Ciaston
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Dobosz
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Kolaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Katarzyna Kjøge
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Maciej Lech
- LMU Hospital, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Barbara Potempa
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan Potempa
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Joanna Koziel
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
15
|
Kim JY, Han HJ. Case report: In vivo detection of neutrophil extracellular traps in a dog with thrombosis induced by bacterial vasculitis. Front Vet Sci 2025; 12:1470605. [PMID: 40012748 PMCID: PMC11862914 DOI: 10.3389/fvets.2025.1470605] [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: 07/25/2024] [Accepted: 01/10/2025] [Indexed: 02/28/2025] Open
Abstract
This case report describes NETosis as a cause of thrombosis in an 18.3 kg, 8-year-old intact male mixed-breed dog with bacterial vasculitis. The dog presented with sudden paresis of the thoracic limb, characterized by cyanosis, absent arterial pulse, and decreased peripheral blood glucose levels. Doppler ultrasound confirmed thrombosis in the dorsal common digital artery. Histopathology post-amputation revealed bacterial vasculitis, thrombosis, and infarction, with immunohistochemical staining identifying extracellular citrullinated histone H3 (CitH3), indicative of NETs involvement. Treatment included antibiotics, pentoxifylline, and anticoagulants, showing transient improvement before disease progression and euthanasia due to respiratory signs. These findings suggest NETs as a potential therapeutic target for bacterial vasculitis in similar cases.
Collapse
Affiliation(s)
| | - Hyun-Jung Han
- Department of Veterinary Emergency and Critical Care, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Setarehaseman A, Mohammadi A, Maitta RW. Thrombocytopenia in Sepsis. Life (Basel) 2025; 15:274. [PMID: 40003683 PMCID: PMC11857489 DOI: 10.3390/life15020274] [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: 12/31/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Platelets, traditionally known for their role in hemostasis, have emerged as key players in immune response and inflammation. Sepsis, a life-threatening condition characterized by systemic inflammation, often presents with thrombocytopenia, which at times, can be significant. Platelets contribute to the inflammatory response by interacting with leukocytes, endothelial cells, and the innate immune system. However, excessive platelet activation and consumption can lead to thrombocytopenia and exacerbate the severity of sepsis. Understanding the multifaceted roles of platelets in sepsis is crucial for developing effective therapeutic strategies. Targeting platelet-mediated inflammatory responses and promoting platelet production may offer potential avenues for improving outcomes in septic patients with thrombocytopenia. Future research should focus on elucidating the mechanisms underlying platelet dysfunction in sepsis and exploring novel therapeutic approaches to optimize platelet function and mitigate inflammation. This review explores the intricate relationship between platelets, inflammation, and thrombosis in the context of sepsis.
Collapse
Affiliation(s)
- Alireza Setarehaseman
- University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Abbas Mohammadi
- Department of Internal Medicine, Valley Health System, Las Vegas, NV 89119, USA;
| | - Robert W. Maitta
- University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| |
Collapse
|
17
|
Li H, Li C, Fu C, Wang Y, Liang T, Wu H, Wu C, Wang C, Sun T, Liu S. Innovative nanoparticle-based approaches for modulating neutrophil extracellular traps in diseases: from mechanisms to therapeutics. J Nanobiotechnology 2025; 23:88. [PMID: 39915767 PMCID: PMC11800495 DOI: 10.1186/s12951-025-03195-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/02/2025] [Indexed: 02/11/2025] Open
Abstract
Neutrophil extracellular traps (NETs) participate in both host defense and the pathogenesis of various diseases, such as infections, thrombosis, and tumors. While they help capture and eliminate pathogens, NETs' excessive or dysregulated formation can lead to tissue damage and disease progression. Therapeutic strategies targeting NET modulation have shown potential, but challenges remain, particularly in achieving precise drug delivery and maintaining drug stability. Nanoparticle (NP)-based drug delivery systems offer innovative solutions for overcoming the limitations of conventional therapies. This review explores the biological mechanisms of NET formation, their interactions with NPs, and the therapeutic applications of NP-based drug delivery systems for modulating NETs. We discuss how NPs can be designed to either promote or inhibit NET formation and provide a comprehensive analysis of their potential in treating NET-related diseases. Additionally, we address the current challenges and future prospects for NP-based therapies in NET research, aiming to bridge the gap between nanotechnology and NET modulation for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Can Li
- Department of Hematology, The Second Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chenxi Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
18
|
Retter A, Singer M, Annane D. "The NET effect": Neutrophil extracellular traps-a potential key component of the dysregulated host immune response in sepsis. Crit Care 2025; 29:59. [PMID: 39905519 DOI: 10.1186/s13054-025-05283-0] [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: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs) as part of a healthy host immune response. NETs physically trap and kill pathogens as well as activating and facilitating crosstalk between immune cells and complement. Excessive or inadequately resolved NETs are implicated in the underlying pathophysiology of sepsis and other inflammatory diseases, including amplification of the inflammatory response and inducing thrombotic complications. Here, we review the growing evidence implicating neutrophils and NETs as central players in the dysregulated host immune response. We discuss potential strategies for modifying NETs to improve patient outcomes and the need for careful patient selection.
Collapse
Affiliation(s)
- Andrew Retter
- Critical Care, Guy's and St Thomas' NHS Foundation Trust, London, UK.
- School of Immunology and Microbial Sciences, King's College, London, UK.
- Volition, London, UK.
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- IHU PROMETHEUS, Comprehensive Sepsis Center, Garches, France
- University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), Garches, France
| |
Collapse
|
19
|
Zhang G, Zhang K. Screening and Identification of Neutrophil Extracellular Trap-related Diagnostic Biomarkers for Pediatric Sepsis by Machine Learning. Inflammation 2025; 48:212-222. [PMID: 38795170 DOI: 10.1007/s10753-024-02059-6] [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: 02/20/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/27/2024]
Abstract
Neutrophil extracellular trap (NET) is released by neutrophils to trap invading pathogens and can lead to dysregulation of immune responses and disease pathogenesis. However, systematic evaluation of NET-related genes (NETRGs) for the diagnosis of pediatric sepsis is still lacking. Three datasets were taken from the Gene Expression Omnibus (GEO) database: GSE13904, GSE26378, and GSE26440. After NETRGs and differentially expressed genes (DEGs) were identified in the GSE26378 dataset, crucial genes were identified by using LASSO regression analysis and random forest analysis on the genes that overlapped in both DEGs and NETRGs. These crucial genes were then employed to build a diagnostic model. The diagnostic model's effectiveness in identifying pediatric sepsis across the three datasets was confirmed through receiver operating characteristic curve (ROC) analysis. In addition, clinical pediatric sepsis samples were collected to measure the expression levels of important genes and evaluate the diagnostic model's performance using qRT-PCR in identifying pediatric sepsis in actual clinical samples. Next, using the CIBERSORT database, the relationship between invading immune cells and diagnostic markers was investigated in more detail. Lastly, to evaluate NET formation, we measured myeloperoxidase (MPO)-DNA complex levels using ELISA. A group of five important genes (MME, BST1, S100A12, FCAR, and ALPL) were found among the 13 DEGs associated with NET formation and used to create a diagnostic model for pediatric sepsis. Across all three cohorts, the sepsis group had consistently elevated expression levels of these five critical genes as compared to the normal group. Area under the curve (AUC) values of 1, 0.932, and 0.966 indicate that the diagnostic model performed exceptionally well in terms of diagnosis. Notably, when applied to the clinical samples, the diagnostic model also showed good diagnostic capacity with an AUC of 0.898, outperforming the effectiveness of traditional inflammatory markers such as PCT, CRP, WBC, and NEU%. Lastly, we discovered that children with high ratings for sepsis also had higher MPO-DNA complex levels. In conclusion, the creation and verification of a five-NETRGs diagnostic model for pediatric sepsis performs better than established markers of inflammation.
Collapse
Affiliation(s)
- Genhao Zhang
- Department of Blood Transfusion, Zhengzhou University First Affiliated Hospital, Zhengzhou, China.
| | - Kai Zhang
- Department of Medical Laboratory, Zhengzhou University Third Affiliated Hospital, Zhengzhou, China
| |
Collapse
|
20
|
Wang Z, Saxena A, Yan W, Uriarte SM, Siqueira R, Li X. The impact of aging on neutrophil functions and the contribution to periodontitis. Int J Oral Sci 2025; 17:10. [PMID: 39819982 PMCID: PMC11739572 DOI: 10.1038/s41368-024-00332-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/21/2024] [Accepted: 10/21/2024] [Indexed: 01/19/2025] Open
Abstract
The increasing aging population and aging-associated diseases have become a global issue for decades. People over 65 show an increased prevalence and greater severity of periodontitis, which poses threats to overall health. Studies have demonstrated a significant association between aging and the dysfunction of neutrophils, critical cells in the early stages of periodontitis, and their crosstalk with macrophages and T and B lymphocytes to establish the periodontal lesion. Neutrophils differentiate and mature in the bone marrow before entering the circulation; during an infection, they are recruited to infected tissues guided by the signal from chemokines and cytokines to eliminate invading pathogens. Neutrophils are crucial in maintaining a balanced response between host and microbes to prevent periodontal diseases in periodontal tissues. The impacts of aging on neutrophils' chemotaxis, anti-microbial function, cell activation, and lifespan result in impaired neutrophil functions and excessive neutrophil activation, which could influence periodontitis course. We summarize the roles of neutrophils in periodontal diseases and the aging-related impacts on neutrophil functional responses. We also explore the underlying mechanisms that can contribute to periodontitis manifestation in aging. This review could help us better understand the pathogenesis of periodontitis, which could offer novel therapeutic targets for periodontitis.
Collapse
Affiliation(s)
- Zi Wang
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anish Saxena
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA
| | - Wenbo Yan
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Rafael Siqueira
- Department of Periodontics, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| | - Xin Li
- Department of Plastic Surgery, Maxillofacial & Oral Health, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Molecular Pathobiology Department, New York University College of Dentistry, New York, NY, USA.
- Comprehensive Cancer Center, University of Virginia, Charlottesville, USA.
| |
Collapse
|
21
|
Englert H, Rangaswamy C, Kullik GA, Divivier M, Göbel J, Hermans‐Borgmeyer I, Borgmeyer U, Mowen KA, Beerens M, Frye M, Mailer RK, Gelderblom M, Stavrou EX, Preston RJS, Schneider SW, Fuchs TA, Renné T. Sepsis-induced NET formation requires MYD88 but is independent of GSDMD and PAD4. FASEB J 2025; 39:e70301. [PMID: 39777764 PMCID: PMC11707982 DOI: 10.1096/fj.202402514r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025]
Abstract
Neutrophils are peripheral blood-circulating leukocytes that play a pivotal role in host defense against bacterial pathogens which upon activation, they release web-like chromatin structures called neutrophil extracellular traps (NETs). Here, we analyzed and compared the importance of myeloid differentiation factor 88 (MYD88), peptidyl arginine deiminase 4 (PAD4), and gasdermin D (GSDMD) for NET formation in vivo following sepsis and neutrophilia challenge. Injection of lipopolysaccharide (LPS)/E. coli or the transgenic expression of granulocyte colony-stimulating factor (G-CSF), each induced NET-mediated lethal vascular occlusions in mice with combined genetic deficiency in Dnase1 and Dnase1l3 (D1/D1l3-/-). In accordance with the signaling of toll-like receptors, Myd88/D1/D1l3-/- animals were protected from the formation of lethal intravascular NETs during septic conditions. However, this protection was not observed during neutrophilia. It was unexpected to find that both Gsdmd/D1/D1l3-/- and Pad4/D1/D1l3-/- mice were fully capable of forming NETs upon LPS/E.coli challenge. Sepsis equally triggered a similar inflammatory response in these mice characterized by formation of DNA-rich thrombi, vessel occlusions, and mortality from pulmonary embolism, compared to D1/D1l3-/- mice. Pharmacologic GSDMD inhibitors did not reduce PMA-stimulated NET formation in ex vivo models either. Similarly, neither Pad4 nor GSDMD deficiency affected intravascular occlusive NET formation upon neutrophilia challenge. The magnitude of NET production, multi-organ damage, and lethality were comparable to those observed in challenged control mice. In conclusion, our data indicate that NET formation during experimental sepsis and neutrophilia is regulated by distinct stimulus-dependent pathways that may be independent of canonical PAD4 and GSDMD.
Collapse
Affiliation(s)
- Hanna Englert
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Giuliano A. Kullik
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mylène Divivier
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Josephine Göbel
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Irm Hermans‐Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Uwe Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Kerri A. Mowen
- Chemical PhysiologyThe Scripps InstituteLa JollaCaliforniaUSA
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Reiner K. Mailer
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mathias Gelderblom
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Evi X. Stavrou
- Medicine Service, Section of Hematology‐OncologyLouis Stokes Veterans Administration Medical CenterClevelandOhioUSA
- Department of Medicine, Hematology and Oncology DivisionCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Roger J. S. Preston
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Stefan W. Schneider
- Department of Dermatology and VenereologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tobias A. Fuchs
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- NeutrolisCambridgeMassachusettsUSA
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
- Center for Thrombosis and Hemostasis (CTH)Johannes Gutenberg University Medical CenterMainzGermany
| |
Collapse
|
22
|
Feješ A, Šebeková K, Borbélyová V. Pathophysiological Role of Neutrophil Extracellular Traps in Diet-Induced Obesity and Metabolic Syndrome in Animal Models. Nutrients 2025; 17:241. [PMID: 39861371 PMCID: PMC11768048 DOI: 10.3390/nu17020241] [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: 12/01/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The global pandemic of obesity poses a serious health, social, and economic burden. Patients living with obesity are at an increased risk of developing noncommunicable diseases or to die prematurely. Obesity is a state of chronic low-grade inflammation. Neutrophils are first to be recruited to sites of inflammation, where they contribute to host defense via phagocytosis, degranulation, and extrusion of neutrophil extracellular traps (NETs). NETs are web-like DNA structures of nuclear or mitochondrial DNA associated with cytosolic antimicrobial proteins. The primary function of NETosis is preventing the dissemination of pathogens. However, neutrophils may occasionally misidentify host molecules as danger-associated molecular patterns, triggering NET formation. This can lead to further recruitment of neutrophils, resulting in propagation and a vicious cycle of persistent systemic inflammation. This scenario may occur when neutrophils infiltrate expanded obese adipose tissue. Thus, NETosis is implicated in the pathophysiology of autoimmune and metabolic disorders, including obesity. This review explores the role of NETosis in obesity and two obesity-associated conditions-hypertension and liver steatosis. With the rising prevalence of obesity driving research into its pathophysiology, particularly through diet-induced obesity models in rodents, we discuss insights gained from both human and animal studies. Additionally, we highlight the potential offered by rodent models and the opportunities presented by genetically modified mouse strains for advancing our understanding of obesity-related inflammation.
Collapse
Affiliation(s)
| | - Katarína Šebeková
- Institute of Molecular Biomedicine, Medical Faculty, Comenius University, 83303 Bratislava, Slovakia; (A.F.); (V.B.)
| | | |
Collapse
|
23
|
Schoenaker JM, Nelson VS, Henderickx JGE, Terveer EM, Jansen AJG, Porcelijn L, Netelenbos T, Schipperus MR, Kapur R. The intestinal flora: The key to unraveling heterogeneity in immune thrombocytopenia? Blood Rev 2025; 69:101252. [PMID: 39672701 DOI: 10.1016/j.blre.2024.101252] [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: 10/16/2024] [Revised: 11/22/2024] [Accepted: 12/01/2024] [Indexed: 12/15/2024]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by enhanced platelet destruction and impaired platelet production, due to a loss of immune tolerance that leads to targeting of platelets and megakaryocytes by glycoprotein-autoantibodies and/or cytotoxic T cells. There is a high degree of heterogeneity in ITP patients signified by unpredictable disease trajectories and treatment responses. Initial studies in humans have identified intestinal microbiota perturbance in ITP. Recently, gut microbial perturbance has been linked to other autoimmune diseases. Based on these findings, we hypothesize that intestinal microbiota may influence ITP pathophysiology through several mechanisms, including induction of platelet-autoantibody production, increasing complement-dependent platelet cytotoxicity, disturbing T cell homeostasis, impairing megakaryocyte function, and increasing platelet-desialylation and -clearance. The pathophysiological heterogeneity of ITP may, at least in part, be attributed to a perturbed intestinal microbiota. Therefore, a better understanding of intestinal microbiota in ITP may result in a more personalized therapeutic approach.
Collapse
Affiliation(s)
- Jente M Schoenaker
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands.
| | - Vivianne S Nelson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Department of Hematology, HagaZiekenhuis, 2545 AA The Hague, the Netherlands; Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Jannie G E Henderickx
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Department of Medical Microbiology, Leiden University Center of Infectious Diseases (LU-CID) Research, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Elisabeth M Terveer
- Department of Medical Microbiology, Leiden University Center of Infectious Diseases (LU-CID) Research, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Netherlands Donor Feces Bank, LUCID Medical Microbiology & Infection Prevention, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - A J Gerard Jansen
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, the Netherlands.
| | - Leendert Porcelijn
- Sanquin Diagnostic Services, Department of Immunohematology Diagnostics, Sanquin, 1066 CX Amsterdam, the Netherlands.
| | - Tanja Netelenbos
- Department of Hematology, HagaZiekenhuis, 2545 AA The Hague, the Netherlands.
| | | | - Rick Kapur
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
24
|
Tu Y, Chen Y, Li X, Wang Y, Fang B, Ren Y, Wang C. Advances in acute COPD exacerbation: clarifying specific immune mechanisms of infectious and noninfectious factors. Ther Adv Respir Dis 2025; 19:17534666241308408. [PMID: 40098281 PMCID: PMC11915264 DOI: 10.1177/17534666241308408] [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] [Indexed: 03/19/2025] Open
Abstract
Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is the main cause of hospitalization and death of patients with chronic obstructive pulmonary disease. This is largely due to bacterial resistance caused by clinical antibiotic abuse and the limited efficacy of current treatment strategies in managing noninfectious AECOPD, which presents a significant challenge for clinicians. Therefore, it is urgent for clinical treatment and prevention of AECOPD to fully understand the specific mechanism of AECOPD in the immune system and master the key differences between infectious factors and noninfectious factors. This article systematically discusses AECOPD triggered by various factors, including the activation of immune system, the recruitment and activation of inflammatory cells and the role of specific inflammatory responses, and through a comprehensive review of the literature, this article expounds the existing targeted diagnosis and treatment methods and technologies at different stages in order to provide new ideas and strategies for clinical prevention and treatment of AECOPD.
Collapse
Affiliation(s)
- Yadan Tu
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Yong Chen
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xuanhan Li
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Yigang Wang
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Bangjiang Fang
- Emergency Department, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Ren
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing 400021, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chenghu Wang
- Department of Classic Chinese Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing, China
- Classic Department of Traditional Chinese Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
25
|
Tong D, Gao Y, Sun W, Yang J, Liu Y, Li J, Zhang Y. Neutrophil Extracellular Traps, Platelets and Endothelial Cells Cooperatively Contribute to Hypercoagulability in Non-Small Cell Lung Cancer. Thromb Haemost 2024. [PMID: 39613309 DOI: 10.1055/a-2493-2499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
BACKGROUND Thromboembolism is the second leading cause of death among patients with non-small cell lung cancer (NSCLC), but the precise mechanisms of thrombogenesis in NSCLC remain largely unknown. Our objectives were to evaluate the definitive role of neutrophil extracellular traps (NETs) in the hypercoagulability in NSCLC and to explore its interactions with platelets and endothelial cells (ECs). METHODS The levels of NET markers in samples from 100 NSCLC patients and 30 healthy controls were measured by ELISA. NET formation was detected using immunofluorescence. Procoagulant activity was assessed based on purified coagulation complex, thrombin, clotting time, and fibrin formation assays. RESULTS The plasma levels of NETs were increased in a stage-dependent manner in NSCLC patients and were markedly higher than those in controls. Neutrophils from NSCLC patients were more prone to form NETs, resulting in shortened coagulation time, significantly increased thrombin-antithrombin complexes and fibrin compared to controls. Moreover, NETs generation was mediated by High Mobility Group Box 1 from activated platelets in NSCLC patients. Conversely, NETs from NSCLC patients also induce phosphatidylserine exposure on platelets, leading to markedly enhanced procoagulant activity (PCA). Furthermore, NETs can damage endothelial cells and convert them to a procoagulant phenotype. The administration of NETs inhibitors (DNase I/activated protein C) could markedly diminish the PCA of NETs, activated platelets, and ECs. CONCLUSION Our results suggest that NETs contribute to hypercoagulability and may represent a potential therapeutic target to prevent cancer-associated thrombosis in NSCLC patients.
Collapse
Affiliation(s)
- Dongxia Tong
- Departments of Oncology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Yuan Gao
- Departments of Gynaecology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Weihua Sun
- Departments of Oncology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Jie Yang
- Departments of Oncology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Yang Liu
- Departments of Oncology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Jihe Li
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| | - Yan Zhang
- Departments of Oncology, Qingdao Municipal Hospital, Qingdao University, Shandong Province, China
| |
Collapse
|
26
|
Zeng F, Shao Y, Wu J, Luo J, Yue Y, Shen Y, Wang Y, Shi Y, Wu D, Cata JP, Yang S, Zhang H, Miao C. Tumor metastasis and recurrence: The role of perioperative NETosis. Cancer Lett 2024; 611:217413. [PMID: 39725150 DOI: 10.1016/j.canlet.2024.217413] [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: 04/29/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Although surgical resection of tumor mass remains the mainstay of curative therapeutic management for solid tumors, accumulating studies suggest that these procedures promote tumor recurrence and metastasis. Regarded as the first immune cells to fight against infectious or inflammatory insults from surgery, neutrophils along with their ability of neutrophil extracellular traps (NETs) production has attracted much attention. A growing body of evidence suggests that NETs promote cancer metastasis by stimulating various stages, including local invasion, colonization, and growth. Therefore, we discussed the mechanism of NETosis induced by surgical stress and tumor cells, and the contribution of NETs on tumor metastasis: aid in the tumor cell migration and proliferation, evasion of immune surveillance, circulating tumor cell adhesion and establishment of a metastatic niche. Lastly, we summarized existing NET-targeting interventions, offering recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Fu Zeng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jingyi Wu
- Department of Anesthesiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jingwen Luo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Dan Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Pujian Road 160, Shanghai, 200127, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
27
|
Grin PM, Baid K, de Jesus HCR, Kozarac N, Bell PA, Jiang SZ, Kappelhoff R, Butler GS, Leborgne NGF, Pan C, Pablos I, Machado Y, Vederas JC, Kim H, Benarafa C, Banerjee A, Overall CM. SARS-CoV-2 3CL pro (main protease) regulates caspase activation of gasdermin-D/E pores leading to secretion and extracellular activity of 3CL pro. Cell Rep 2024; 43:115080. [PMID: 39673710 DOI: 10.1016/j.celrep.2024.115080] [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: 06/13/2024] [Revised: 10/27/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024] Open
Abstract
SARS-CoV-2 3C-like protease (3CLpro or Mpro) cleaves the SARS-CoV-2 polyprotein and >300 intracellular host proteins to enhance viral replication. By lytic cell death following gasdermin (GSDM) pore formation in cell membranes, antiviral pyroptosis decreases 3CLpro expression and viral replication. Unexpectedly, 3CLpro and nucleocapsid proteins undergo unconventional secretion from infected cells via caspase-activated GSDMD/E pores in the absence of cell lysis. Bronchoalveolar lavage fluid of wild-type SARS-CoV-2-infected mice contains 3CLpro, which decreases in Gsdmd-/-Gsdme-/- mice. We identify new 3CLpro cut-sites in GSDMD at LQ29↓30SS, which blocks pore formation by 3CLpro cleavage at LH270↓N lying adjacent to the caspase activation site (NFLTD275↓G). Cleavage inactivation of GSDMD prevents excessive pore formation, thus countering antiviral pyroptosis and increasing 3CLpro secretion. Extracellular 3CLpro retains activity in serum, dampens platelet activation and aggregation, and inactivates antiviral interferon-λ1. Thus, in countering gasdermin pore formation and pyroptosis in SARS-CoV-2 infection, 3CLpro is secreted with extracellular pathological sequelae.
Collapse
Affiliation(s)
- Peter M Grin
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Kaushal Baid
- Vaccine and Infectious Diseases Organization, Department of Veterinary Microbiology, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada
| | - Hugo C R de Jesus
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nedim Kozarac
- Institute for Virology and Immunology IVI, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Peter A Bell
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Steven Z Jiang
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Reinhild Kappelhoff
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Georgina S Butler
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nathan G F Leborgne
- Institute for Virology and Immunology IVI, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland
| | - Christina Pan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Isabel Pablos
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Yoan Machado
- Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - John C Vederas
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr. NW, Edmonton, AB T6G 2N4, Canada
| | - Hugh Kim
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Charaf Benarafa
- Department of Infectious Diseases and Pathology, Vetsuisse Faculty, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland; Multidisciplinary Center for Infectious Diseases, University of Bern, Sensemattstrasse 293, 3147 Mittelhäusern, Switzerland.
| | - Arinjay Banerjee
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Vaccine and Infectious Diseases Organization, Department of Veterinary Microbiology, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada; Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Simcoe Hall, 1 King's College Cir., Toronto, ON M5S 1A8, Canada.
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Centre for Blood Research, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Yonsei Frontier Lab, Yonsei University, 50 Yonsei-ro, Sudaemoon-ku, Seoul 03722, Republic of Korea.
| |
Collapse
|
28
|
Ono M, Toyomoto M, Yamauchi M, Hagiwara M. Platelets accelerate lipid peroxidation and induce pathogenic neutrophil extracellular trap release. Cell Chem Biol 2024; 31:2085-2095.e4. [PMID: 39631397 DOI: 10.1016/j.chembiol.2024.11.003] [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: 09/11/2023] [Revised: 08/02/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Neutrophil extracellular traps (NETs), an important host defense mechanism, are assembled after the release of decondensed chromatin and other nuclear components by a process termed NETosis. However, excessive NET release destroys surrounding tissues, leading to conditions such as sepsis where platelets are implicated in the pathogenic switch of NETosis. Here, we show that platelets trigger iron accumulation and promote lipid peroxide production in neutrophils co-stimulated with lipopolysaccharide and platelets in vitro, resulting in the induction of NETosis. We also screened for compounds that inhibit lipid peroxidation, identified 8-methyl-N-geranyl-6-nonamide (capsaicin), and assessed its potential in suppressing platelet-mediated pathogenic NETosis. Capsaicin inhibited lipopolysaccharide/platelet-induced cellular lipid peroxidation and suppressed NETosis in vitro. Furthermore, capsaicin attenuated NETosis in a mouse model of lipopolysaccharide-induced lung inflammation. Our findings provide an original therapeutic strategy to target lipid peroxidation and pave the way for drug development for a wide range of NETosis-related diseases.
Collapse
Affiliation(s)
- Madoka Ono
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masayasu Toyomoto
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Department of Drug Discovery for Intractable Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Momono Yamauchi
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
29
|
Chowdhury CS, Kinsella RL, McNehlan ME, Naik SK, Lane DS, Talukdar P, Smirnov A, Dubey N, Rankin AN, McKee SR, Woodson R, Hii A, Chavez SM, Kreamalmeyer D, Beatty W, Mattila JT, Stallings CL. Type I IFN-mediated NET release promotes Mycobacterium tuberculosis replication and is associated with granuloma caseation. Cell Host Microbe 2024; 32:2092-2111.e7. [PMID: 39637864 PMCID: PMC11637906 DOI: 10.1016/j.chom.2024.11.008] [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: 12/13/2022] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Neutrophils are the most abundant cell type in the airways of tuberculosis patients. Mycobacterium tuberculosis (Mtb) infection induces the release of neutrophil extracellular traps (NETs); however, the molecular regulation and impact of NET release on Mtb pathogenesis are unknown. We find that during Mtb infection in neutrophils, PAD4 citrullinates histones to decondense chromatin that gets released as NETs in a manner that can maintain neutrophil viability and promote Mtb replication. Type I interferon promotes the formation of chromatin-containing vesicles that allow NET release without compromising plasma membrane integrity. Analysis of nonhuman primate granulomas supports a model where neutrophils are exposed to type I interferon from macrophages as they migrate into the granuloma, thereby enabling the release of NETs associated with necrosis and caseation. Our data reveal NET release as a promising target to inhibit Mtb pathogenesis.
Collapse
Affiliation(s)
- Chanchal Sur Chowdhury
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Kinsella
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael E McNehlan
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sumanta K Naik
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel S Lane
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Priyanka Talukdar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Asya Smirnov
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Neha Dubey
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ananda N Rankin
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel R McKee
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reilly Woodson
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Abigail Hii
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA; UCD School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Sthefany M Chavez
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Darren Kreamalmeyer
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
30
|
Nakagawa R, Itokazu T, Shibuya N, Kishima H, Yamashita T. Perivascular Neutrophil Extracellular Traps Exacerbate Microvasospasm After Experimental Subarachnoid Hemorrhage. Stroke 2024; 55:2872-2881. [PMID: 39474691 DOI: 10.1161/strokeaha.124.047574] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/25/2024] [Accepted: 09/11/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) can lead to acute or delayed cerebral ischemia. Recent findings have revealed that spasm of microvessels, called microvasospasm, may contribute to SAH-related cerebral ischemia, and perivascular inflammation is considered important in the development of microvasospasms. However, owing to the difficulty in investigating the dynamics of vascular and perivascular events, little is known about the mechanisms underlying microvasospasms. METHODS We established an experimental system aiming to investigate the vascular and perivascular pathology of SAH by combining a SAH mouse model with intravital 2-photon imaging. SAH was induced by intracisternal blood injection, and the distribution of erythrocytes, neutrophil behavior, and morphological changes in the pial arterioles were analyzed over time by 2-photon microscopy imaging. To further explore the role of neutrophils and neutrophil extracellular traps (NETs) in microvasospasm, we performed neutrophil depletion by intraperitoneal administration of neutrophil-specific antibody or NETs removal by intracisternal administration of DNase. RESULTS Erythrocytes were immediately distributed in the perivascular space of the arterioles after SAH induction; neutrophils intensively infiltrated the perivascular space within 2 days and subsequently showed NETosis; and pial arterioles in the same region developed pearl-string-like microvasospasms in the subacute phase. Neutrophil depletion significantly reduced the number of microvasospasms. Furthermore, the removal of perivascular NETs drastically reduced microvasospasms. CONCLUSIONS By establishing a unique experimental system, we demonstrated that perivascular NETs could be a new therapeutic target for microvasospasms.
Collapse
Affiliation(s)
- Ryota Nakagawa
- Department of Molecular Neuroscience (R.N., T.I., N.S., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Neurosurgery (R.N., H.K.), Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience (R.N., T.I., N.S., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Neuro-Medical Science (T.I., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
| | - Nao Shibuya
- Department of Molecular Neuroscience (R.N., T.I., N.S., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery (R.N., H.K.), Graduate School of Medicine, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience (R.N., T.I., N.S., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Neuro-Medical Science (T.I., T.Y.), Graduate School of Medicine, Osaka University, Suita, Japan
- WPI Immunology Frontier Research Center, Osaka, Japan (T.Y.)
- Graduate School of Frontier Biosciences, Osaka University, Japan (T.Y.)
| |
Collapse
|
31
|
Wang N, Shi XL, Li D, Li BB, Liu P, Luo H. Neutrophil extracellular traps - an a-list-actor in a variety of diseases. Ann Hematol 2024; 103:5059-5069. [PMID: 39078437 DOI: 10.1007/s00277-024-05915-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024]
Abstract
Neutrophil extracellular traps (NETs) represent a response mechanism in which activated neutrophils release DNA-based webs, adorned with histones and neutrophil proteases, to capture and eliminate invasive microorganisms. However, when these neutrophils become excessively activated, much more proteases associated with NETs are liberated into surrounding tissues or bloodstreams, thereby altering the cellular milieu and causing tissue damage. Recent research has revealed that NETs may play significant roles in the emergence and progression of various diseases, spanning from infections, inflammation to autoimmune disorders and cancers. In this review, we delve deeply into the intricate and complex mechanisms that underlie the formation of NETs and their profound interplay with various clinical pathologies. We aim to describe the application perspectives of NETs related proteins in specific disease diagnosis and treatment.
Collapse
Affiliation(s)
- Na Wang
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning, 116044, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Xiao-Lin Shi
- Department of Clinical Laboratory, Weihai Maternal and Child Health Hospital, Weihai, Shandong, 264200, PR China
| | - Dan Li
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Bin-Bin Li
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, PR China.
| | - Hong Luo
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning, 116044, PR China.
| |
Collapse
|
32
|
George Pryzdial EL, Perrier JR, Rashid MU, West HE, Sutherland MR. Viral coagulation: pushing the envelope. J Thromb Haemost 2024; 22:3366-3382. [PMID: 39260743 DOI: 10.1016/j.jtha.2024.08.014] [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: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Many virus types affect the blood clotting system with correlations to pathology that range widely from thrombosis to hemorrhage linking to inflammation. Here we overview the intricate crosstalk induced by infection between proteins on the virus encoded by either the host or virus genomes, coagulation proteins, platelets, leukocytes, and endothelial cells. For blood-borne viruses with an outer covering acquired from the host cell, the envelope, a key player may be the cell-derived trigger of coagulation on the virus surface, tissue factor (TF). TF is a multifunctional transmembrane cofactor that accelerates factor (F)VIIa-dependent activation of FX to FXa, leading to clot formation. However, the nascent TF/FVIIa/FXa complex also facilitates G protein-coupled modulation of cells via protease-activated receptor 2. As a viral envelope constituent, TF can bypass the physiological modes of regulation, thereby initiating the activation of neighboring platelets, leukocytes, and endothelial cells. A thromboinflammatory environment is predicted due to feedback amplification in response to cellular release of cytokines, procoagulant proteins, neutrophil extracellular traps, and stimulus-induced accessibility of adhesive receptors, resulting in cellular aggregates. The pathobiological effects of thromboinflammation ultimately contribute to innate and adaptive immunity for viral clearance. In contrast, the preceding stages of viral infection may be enhanced via the TF-protease axis.
Collapse
Affiliation(s)
- Edward Louis George Pryzdial
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.
| | - John Ruggles Perrier
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mahamud-Ur Rashid
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Henry Euan West
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Michael Ross Sutherland
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Maugeri N, Manfredi AA. Platelet HMGB1 steers intravascular immunity and thrombosis. J Thromb Haemost 2024; 22:3336-3345. [PMID: 39173879 DOI: 10.1016/j.jtha.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Platelets navigate the fine balance between homeostasis and injury. They regulate vascular homeostasis and drive repair after injury amidst leukocyte extravasation. Crucially, platelets initiate extracellular traps generation and promote immunothrombosis. In chronic human diseases, platelet action often extends beyond its normative role, sparking sustained reciprocal activation of leukocytes and mural cells, culminating in adverse vascular remodeling. Studies in the last decade have spotlighted a novel key player in platelet activation, the high mobility group box 1 (HMGB1) protein. Despite its initial characterization as a chromatin molecule, anucleated platelets express abundant HMGB1, which has emerged as a linchpin in thromboinflammatory risks and microvascular remodeling. We propose that a comprehensive assessment of platelet HMGB1, spanning quantification of content, membrane localization, and accumulation of HMGB1-expressing vesicles in biological fluids should be integral to dissecting and quantifying platelet activation. This review provides evidence supporting this claim and underscores the significance of platelet HMGB1 as a biomarker in conditions associated with heightened thrombotic risks and systemic microvascular involvement, spanning cardiovascular, autoimmune, and infectious diseases.
Collapse
Affiliation(s)
- Norma Maugeri
- Division of Immunology, Transplantation & Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico San Raffaele Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy.
| | - Angelo A Manfredi
- Division of Immunology, Transplantation & Infectious Diseases, Istituti di Ricovero e Cura a Carattere Scientifico San Raffaele Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
34
|
Wu Y, Shen J. Unraveling the intricacies of neutrophil extracellular traps in inflammatory bowel disease: Pathways, biomarkers, and promising therapies. Cytokine Growth Factor Rev 2024; 80:156-167. [PMID: 39438227 DOI: 10.1016/j.cytogfr.2024.10.003] [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: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
The development of inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, involves various factors and is characterized by persistent inflammation of the mucosal lining. However, the role of neutrophils in this process remains controversial. Neutrophil extracellular traps (NETs), which consist of chromatin, antimicrobial proteins, and oxidative enzymes, are released by neutrophils to trap pathogens. They are also involved in various immune-mediated and vascular diseases. NETs act as a vital defense mechanisms at the gut-mucosal interface and are frequently exposed to bacterial, viral, and fungal threats. However, they can also contribute to inflammation and worsen imbalances in the gut bacteria. Recent studies have suggested that NETs have a significant impact on IBD development. Previous studies have shown increased levels of NETs in tissue and blood samples from patients with IBD, as well as in experimental colitis mouse models. Therefore, this review discusses how NETs are formed and their role in the pathophysiology of IBD. It discusses how NETs may lead to tissue damage and contribute to IBD-associated complications. Moreover, non-invasive biomarkers are needed to replace invasive procedures such as endoscopy to better evaluate the disease status. Given the crucial role of NETs in IBD progression, this review focuses on potential NET biomarkers that can help predict the evolution of IBD. Furthermore, this review identifies potential therapeutic targets for regulating NET production, which could expand the range of available treatment options for IBD.
Collapse
Affiliation(s)
- Yilin Wu
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China.
| |
Collapse
|
35
|
Tokarz-Deptuła B, Baraniecki Ł, Palma J, Stosik M, Deptuła W. Characterization of Platelet Receptors and Their Involvement in Immune Activation of These Cells. Int J Mol Sci 2024; 25:12611. [PMID: 39684330 DOI: 10.3390/ijms252312611] [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: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The article characterises platelets, pointing out the role and contribution of their numerous receptors determining their specific and broad immune activity. Three types of platelet receptors are described, that is, extracellular and intracellular receptors-TLR (toll-like receptors), NLR (NOD-like receptor), and RLR (RIG-I-like receptor); extracellular receptors-selectins and integrins; and their other extracellular receptors-CLR (C-type lectin receptor), CD (cluster of differentiation), TNF (tumour necrosis factor), among others. Outlining the contribution of these numerous platelet receptors to the intravascular immunity, it has been shown that they are formed by their fusion with pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and lifestyle-associated molecular patterns (LAMPs). They are initiating and effector components of signal transduction of these cells, and their expression and quantity determine the specific and broad functions of platelets towards influencing vascular endothelial cells, but mainly PRRs (pattern recognition receptors) of blood immune cells. These facts make platelets the fundamental elements that shape not only intravascular homeostasis, as previously indicated, but they become the determinants of immunity in blood vessels. Describing the reactions of the characterised three groups of platelet receptors with PAMP, DAMP and LAMP molecules, the pathways and participation of platelets in the formation and construction of intravascular immune status, in physiological states, but mainly in pathological states, including bacterial and viral infections, are presented, making these cells essential elements in the health and disease of mammals, including humans.
Collapse
Affiliation(s)
| | - Łukasz Baraniecki
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Doctoral School, University of Szczecin, 70-384 Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, 65-516 Zielona Góra, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| |
Collapse
|
36
|
Bayram YE, Bardakci MI, Albayrak GA. Improved kidney function is associated with Colchicine treatment in COVID-19 patients. BMC Nephrol 2024; 25:405. [PMID: 39529013 PMCID: PMC11556156 DOI: 10.1186/s12882-024-03817-2] [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: 07/08/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus) has been a major cause of significant morbidity and mortality. Acute kidney injury (AKI) has been seen in COVID-19-infected subjects, and it has frequently resulted in an abnormal estimated glomerular filtration rate. Colchicine, an immunomodulatory drug, was used in several studies in the early stages of the pandemic. Colchicine has been shown to prevent the development of renal failure in patients with Familial Mediterranean Fever (FMF). It has also been reported to reduce fibrosis, which plays a role in chronic kidney disease. We, therefore, aimed to investigate whether using Colchicine, in addition to standard care, was associated with better renal function in patients with severe COVID-19 infection. METHODS This retrospective cohort study comprised 118 out of 605 hospitalized COVID-19 subjects. Some of the subjects (n = 50) received oral Colchicine plus standard care, called the Col ( +) group. The others (n = 68) received only the standard care, called the Col (-) group. The estimated glomerular filtration rate (eGFR) and other laboratory findings, including lymphocytes, D-dimer, and CRP, were analyzed. RESULTS The D-dimer and serum creatine levels were significantly reduced in both groups. The number of lymphocytes showed a significant increase in both groups at discharge. The level of C-reactive protein (CRP) was significantly higher in the Col ( +) group than in the Col (-) group at admission. The reduction of SCr was considerably higher in the Col ( +) group than in the Col (-) group. Similarly, the improvement of eGFR was higher in the Col ( +) group than in the Col (-) group at discharge and 6-12 mounts follow-up. CONCLUSION Our findings indicated the use of Colchicine plus standard care was associated with improved renal function in hospitalized patients with severe COVID-19 infection.
Collapse
Affiliation(s)
- Yeter Eylul Bayram
- Department of Internal Medicine, Sisli Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey.
| | - Mustafa Ilteris Bardakci
- Department of Pulmonology, Sisli Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey
| | - Gulhan Ayhan Albayrak
- Department of Pulmonology, Sisli Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey
| |
Collapse
|
37
|
Yu X, Chen Z, Bao W, Jiang Y, Ruan F, Wu D, Le K. The neutrophil extracellular traps in neurological diseases: an update. Clin Exp Immunol 2024; 218:264-274. [PMID: 38975702 PMCID: PMC11557138 DOI: 10.1093/cei/uxae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/08/2024] [Accepted: 07/06/2024] [Indexed: 07/09/2024] Open
Abstract
Neutrophil extracellular traps (NETs) released by neutrophils are web-like DNA structures adhered to granulin proteins with bactericidal activity and can be an important mechanism for preventing pathogen dissemination or eliminating microorganisms. However, they also play important roles in diseases of other systems, such as the central nervous system. We tracked the latest advances and performed a review based on published original and review articles related to NETs and neurological diseases. Generally, neutrophils barely penetrate the blood-brain barrier into the brain parenchyma, but when pathological changes such as infection, trauma, or neurodegeneration occur, neutrophils rapidly infiltrate the central nervous system to exert their defensive effects. However, neutrophils may adversely affect the host when they uncontrollably release NETs upon persistent neuroinflammation. This review focused on recent advances in understanding the mechanisms and effects of NETs release in neurological diseases, and we also discuss the role of molecules that regulate NETs release in anticipation of clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong S.A.R., China
| |
Collapse
|
38
|
Johansson K, Maouia A, Rebetz J, Marcoux G, Shannon O, Italiano JE, Narayanan P, Henry S, Shen L, Semple JW. CpG oligonucleotides induce acute murine thrombocytopenia dependent on toll-like receptor 9 and spleen tyrosine kinase pathways. J Thromb Haemost 2024; 22:3266-3276. [PMID: 39155024 DOI: 10.1016/j.jtha.2024.08.003] [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: 07/12/2023] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND CpG oligonucleotides (ODNs) are synthetic single-stranded DNA sequences that act as immunostimulants. They have been increasingly used to treat several cancers; however, thrombocytopenia is a potential recognized side effect of some sequences. OBJECTIVES We tested the ability of 2 CpG ODNs (ODN 2395 and ISIS 120704) to induce thrombocytopenia when administered to BALB/c mice and determined mechanisms associated with thrombocytopenia. METHODS BALB/c mice were prebled and then injected with titrated doses of CpG ODNs, and platelet counts were determined. The mice were treated with intravenous immunoglobulin (IVIg) or various inhibitors and antagonists of toll-like receptor 9 (TLR9) and spleen tyrosine kinase (Syk) to determine their effects on thrombocytopenia. RESULTS Compared with saline-treated mice or mice treated with 2'-O-methoxyethyl-modified antisense ODN, both ODN 2395 and ISIS 120704 induced acute dose-dependent thrombocytopenia within 3 and 24 hours, respectively. The thrombocytopenia was associated with significant increases in plasma monocyte chemoattractant protein 1. IVIg administration significantly rescued the CpG ODN-induced thrombocytopenia, as did treatment with either a Syk inhibitor or TLR9 antagonists. In vitro, CpG ODN could activate human platelets and this correlated significantly with enhanced IVIg- and Syk-dependent phagocytosis by THP-1 monocytes. CONCLUSION These results suggest that CpG ODNs induce acute inflammatory-associated (IVIg-sensitive) thrombocytopenia that can be alleviated by Syk- or TLR9-blockade, and an IVIg- and Syk-dependent platelet clearance pathway appears primarily responsible for the thrombocytopenia.
Collapse
Affiliation(s)
- Karl Johansson
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Amal Maouia
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Geneviève Marcoux
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Oonagh Shannon
- Division of Infection Medicine, Lund University, Lund, Sweden
| | - Joseph E Italiano
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Scott Henry
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Lijiang Shen
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden.
| |
Collapse
|
39
|
Argueta DA, Tran H, Goel Y, Nguyen A, Nguyen J, Kiven SB, Chen C, Abdulla F, Vercellotti GM, Belcher JD, Gupta K. Mast cell extracellular trap formation underlies vascular and neural injury and hyperalgesia in sickle cell disease. Life Sci Alliance 2024; 7:e202402788. [PMID: 39242155 PMCID: PMC11381676 DOI: 10.26508/lsa.202402788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Sickle cell disease (SCD) is the most common inherited monogenetic disorder. Chronic and acute pain are hallmark features of SCD involving neural and vascular injury and inflammation. Mast cells reside in the vicinity of nerve fibers and vasculature, but how they influence these structures remains unknown. We therefore examined the mechanism of mast cell activation in a sickle microenvironment replete with cell-free heme and inflammation. Mast cells exposed to this environment showed an explosion of nuclear contents with the release of citrullinated histones, suggestive of mast cell extracellular trap (MCET) release. MCETs interacted directly with the vasculature and nerve fibers, a cause of vascular and neural injury in sickle cell mice. MCET formation was dependent upon peptidylarginine deiminase 4 (PAD4). Inhibition of PAD4 ameliorated vasoocclusion, chronic and acute hyperalgesia, and inflammation in sickle mice. PAD4 activation may also underlie neutrophil trap formation in SCD, thus providing a novel target to treat the sequelae of vascular and neural injury in SCD.
Collapse
Affiliation(s)
- Donovan A Argueta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Huy Tran
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Yugal Goel
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Aithanh Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Julia Nguyen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Stacy B Kiven
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Chunsheng Chen
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Fuad Abdulla
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - John D Belcher
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Kalpna Gupta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
- Division of Hematology, Oncology, and Transplantation, School of Medicine, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
40
|
Li S, Xu G, Guo Z, Liu Y, Ouyang Z, Li Y, Huang Y, Sun Q, Giri BR, Fu Q. Deficiency of hasB accelerated the clearance of Streptococcus equi subsp. Zooepidemicus through gasdermin d-dependent neutrophil extracellular traps. Int Immunopharmacol 2024; 140:112829. [PMID: 39083933 DOI: 10.1016/j.intimp.2024.112829] [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: 04/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Streptococcus equi subsp. zooepidemicus (S. zooepidemicus, SEZ) is an essential zoonotic bacterial pathogen that can cause various inflammation, such as meningitis, endocarditis, and pneumonia. UDP-glucose dehydrogenase (hasB) is indispensable in synthesizing SEZ virulence factor hyaluronan capsules. Our study investigated the infection of hasB on mice response to SEZ by employing a constructed capsule-deficient mutant strain designated as the ΔhasB strain. This deficiency was associated with a reduced SEZ bacterial load in the mice's blood and peritoneal lavage fluid (PLF) post-infection. Besides, the ΔhasB SEZ strain exhibited a higher propensity for neutrophil infiltration and release of cell-free DNA (cfDNA) in vivo compared to the wild-type (WT) SEZ strain. In vitro experiments further revealed that ΔhasB SEZ more effectively induced the formation of neutrophil extracellular traps (NETs) containing histone 3 (H3), neutrophil elastase (NE), and DNA, than its WT counterpart. Moreover, the release of NETs was determined to be gasdermin D (GSDMD)-dependent during the infection process. Taken together, these findings underscore that the deficiency of the hasB gene in SEZ leads to enhanced GSDMD-dependent NET release from neutrophils, thereby reducing SEZ's capacity to resist NETs-mediated eradication during infection. Our finding paves the way for the development of innovative therapeutic strategies against SEZ.
Collapse
Affiliation(s)
- Shun Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Guobin Xu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zheng Guo
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Yuxuan Liu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhiliang Ouyang
- Houjie Town Agricultural Technology Service Center, Dongguan, Guangdong, China
| | - Yajuan Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Qinqin Sun
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China
| | - Bikash R Giri
- Department of Zoology, K.K.S. Women's College, Balasore, Odisha, India
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China; Foshan University Veterinary Teaching Hospital, Foshan University, Foshan, Guangdong, China.
| |
Collapse
|
41
|
Wu G, Pan B, Shi H, Yi Y, Zheng X, Ma H, Zhao M, Zhang Z, Cheng L, Huang Y, Guo W. Neutrophils' dual role in cancer: from tumor progression to immunotherapeutic potential. Int Immunopharmacol 2024; 140:112788. [PMID: 39083923 DOI: 10.1016/j.intimp.2024.112788] [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: 02/01/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
The tumor microenvironment (TME) is intricately associated with cancer progression, characterized by dynamic interactions among various cellular and molecular components that significantly impact the carcinogenic process. Notably, neutrophils play a crucial dual role in regulating this complex environment. These cells oscillate between promoting and inhibiting tumor activity, responding to a multitude of cytokines, chemokines, and tumor-derived factors. This response modulates immune reactions and affects the proliferation, metastasis, and angiogenesis of cancer cells. A significant aspect of their influence is their interaction with the endoplasmic reticulum (ER) stress responses in cancer cells, markedly altering tumor immunodynamics by modulating the phenotypic plasticity and functionality of neutrophils. Furthermore, neutrophil extracellular traps (NETs) exert a pivotal influence in the progression of malignancies by enhancing inflammation, metastasis, immune suppression, and thrombosis, thereby exacerbating the disease. In the realm of immunotherapy, checkpoint inhibitors targeting PD-L1/PD-1 and CTLA-4 among others have underscored the significant role of neutrophils in enhancing therapeutic responses. Recent research has highlighted the potential of using neutrophils for targeted drug delivery through nanoparticle systems, which precisely control drug release and significantly enhance antitumor efficacy. This review thoroughly examines the diverse functions of neutrophils in cancer treatment, emphasizing their potential in regulating immune therapy responses and as drug delivery carriers, offering innovative perspectives and profound implications for the development of targeted diagnostic and therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaobin Zheng
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, China
| | - Lin Cheng
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI), Galway, Ireland.
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weigang Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Fu Y, He Y, Zheng C, Zeng J, Ou H. A Predictive Model for Disseminated Intravascular Coagulopathy in Sepsis: An Observational Study. Int J Gen Med 2024; 17:4845-4855. [PMID: 39465188 PMCID: PMC11512532 DOI: 10.2147/ijgm.s475953] [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: 07/17/2024] [Accepted: 10/06/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Sepsis remains a significant global health challenge due to its high morbidity and mortality rates. Disseminated Intravascular Coagulopathy (DIC) represents a critical complication of sepsis, contributing to increased mortality and economic burden. Despite various prognostic scoring systems, there is a lack of a specific model for DIC prediction in sepsis patients. Methods This observational study included 336 sepsis patients. Clinical and laboratory data were collected, and prognoses were defined according to established criteria. Results We enrolled 336 patients, with 304 in the non-DIC group and 32 in the DIC group. Patients with DIC had notably lower platelet (PLT) and higher levels of prothrombin time (PT), lactate (LAC), and procalcitonin (PCT) compared to those without DIC. Univariate and multivariate analyses identified risk factors associated with the DIC, showing that PLT (OR = 0.985, 95% CI 0.978-0.993, p < 0.001), PT level (OR = 1.140, 95% CI 1.004-1.295, p = 0.044), and LAC (OR = 1.101, 95% CI 0.989-1.226, p = 0.078) were related factors. A risk model was established, and its sensitivity and specificity in predicting DIC among sepsis patients were assessed by comparing it to the SOFA score. The area under the ROC curve for the model was 0.850, while the SOFA score was 0.813. With a model score >-2.12, the sensitivity for predicting DIC was 84.4%, and the specificity was 75.0%. Conclusion Our study introduces a predictive model for DIC detection in sepsis patients, emphasizing the need for clinicians to focus on patients with high model scores for timely intervention.
Collapse
Affiliation(s)
- Yaojie Fu
- Emergency Department, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People’s Republic of China
| | - Yujing He
- Intense Care Unit, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People’s Republic of China
| | - Caixia Zheng
- Department of Infectious Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People’s Republic of China
| | - Jianyong Zeng
- Department of Infectious Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People’s Republic of China
| | - Hongjie Ou
- Department of Infectious Diseases, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People’s Republic of China
| |
Collapse
|
43
|
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
|
44
|
Cross AL, Wright HL, Choi J, Edwards SW, Ruiz-Opazo N, Herrera VLM. Circulating neutrophil extracellular trap-forming neutrophils in rheumatoid arthritis exacerbation are majority dual endothelin-1/signal peptide receptor+ subtype. Clin Exp Immunol 2024; 218:163-168. [PMID: 39110036 PMCID: PMC11482496 DOI: 10.1093/cei/uxae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 08/06/2024] [Indexed: 10/18/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are associated with rheumatoid arthritis pathogenesis and severity. Since homeostatic NET-forming neutrophils [NET+Ns] have beneficial roles in defense against pathogens, their distinction from pro-injury [NET+N] subtypes is important, especially if they are to be therapeutically targeted. Having identified circulating, pro-injury DEspR+CD11b+[NET+Ns] in patients with neutrophilic secondary tissue injury, we determined whether DEspR+[NET+Ns] are present in rheumatoid arthritis (RA) flares. Whole blood samples of patients with RA flares on maintenance therapy (n = 6) were analyzed by flow cytometry (FCM) and immunofluorescence cytology followed by semi-automated quantitative confocal microscopy (qIFC). We assessed clinical parameters, levels of neutrophils and [NET+Ns], and plasma S100A8/A9. qIFC detected circulating DEspR+CD11b+neutrophils and [NET+Ns] in RA-flare patients but not healthy controls. DEspR+[NET+Ns] were positive for citrullinated histone H3 (citH3+), extruded DNA, decondensed but recognizable polymorphic nuclei, and [NET+N] doublet interactions in mostly non-ruptured NET-forming neutrophils. Circulating DNA+/DEspR+/CD11b+/citH3+microvesicles (netMVs) were observed. FCM detected increased %DEspR+CD11b+neutrophils and DEspR+ cell-cell doublets whose levels trended with DAS28 scores, as did plasma S100A8/A9 levels. This study identifies circulating DEspR+/CD11b+neutrophils and [NET+Ns] in RA-flare patients on maintenance therapy. Detection of circulating DEspR+citH3+[NET+Ns] and netMVs indicate a systemic neutrophilic source of citH3-antigen concordant with multi-joint RA pathogenesis. Increased S100A8/A9 alarmin levels are associated with cell injury and released upon NET-formation. As a ligand for TLR4, S100A8/A9 forms a positive feedback loop for TLR4-induced DEspR+neutrophils. These data identify DEspR+neutrophils and [NET+Ns] in RA pathogenesis as a potential biomarker and/or therapeutic target.
Collapse
Affiliation(s)
- Andrew L Cross
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen L Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Jacqueline Choi
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Steven W Edwards
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Nelson Ruiz-Opazo
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Victoria L M Herrera
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
45
|
Xu C, Jing W, Liu C, Yuan B, Zhang X, Liu L, Zhang F, Chen P, Liu Q, Wang H, Du X. Cytoplasmic DNA and AIM2 inflammasome in RA: where they come from and where they go? Front Immunol 2024; 15:1343325. [PMID: 39450183 PMCID: PMC11499118 DOI: 10.3389/fimmu.2024.1343325] [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: 11/23/2023] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease of undetermined etiology characterized by symmetric synovitis with predominantly destructive and multiple joint inflammation. Cytoplasmic DNA sensors that recognize protein molecules that are not themselves or abnormal dsDNA fragments play an integral role in the generation and perpetuation of autoimmune diseases by activating different signaling pathways and triggering innate immune signaling pathways and host defenses. Among them, melanoma deficiency factor 2 (AIM2) recognizes damaged DNA and double-stranded DNA and binds to them to further assemble inflammasome, initiating the innate immune response and participating in the pathophysiological process of rheumatoid arthritis. In this article, we review the research progress on the source of cytoplasmic DNA, the mechanism of assembly and activation of AIM2 inflammasome, and the related roles of other cytoplasmic DNA sensors in rheumatoid arthritis.
Collapse
Affiliation(s)
- Conghui Xu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Zheng's Acupuncture, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Fengfan Zhang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Qiang Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
46
|
Herre M, Vemuri K, Cedervall J, Nissl S, Saupe F, Micallef J, Lindman H, Maguire CA, Tetz G, Tetz V, Olsson A. AAV-mouse DNase I sustains long-term DNase I expression in vivo and suppresses breast cancer metastasis. FASEB Bioadv 2024; 6:454-466. [PMID: 39372124 PMCID: PMC11452440 DOI: 10.1096/fba.2024-00114] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
Neutrophil extracellular traps (NETs) have been implicated in the pathology of various inflammatory conditions. In cancer, NETs have been demonstrated to induce systemic inflammation, impair peripheral vessel and organ function and promote metastasis. Here we show that the plasma level of NETs is significantly higher in patients with metastatic breast cancer compared to those with local disease, or those that were considered cured at a 5-year follow-up, confirming NETs as interesting therapeutic targets in metastatic breast cancer. Administration of DNase I is one strategy to eliminate NETs but long-term treatment requires repeated injections and species-specific versions of the enzyme. To enhance administration and therapeutic efficacy, we have developed an adeno-associated virus (AAV) vector system for delivery of murine DNase I and addressed its potential to counteract cancer-associated pathology in the murine MMTV-PyMT model for metastatic mammary carcinoma. The AAV vector is comprised of capsid KP1 and an expression cassette encoding hyperactive murine DNase I (AAV-mDNase I) under the control of a liver-specific promotor. This AAV-mDNase I vector could support elevated expression and serum activity of murine DNase I over at least 8 months. Neutrophil Gelatinase-Associated Lipocalin (NGAL), a biomarker for kidney hypoperfusion that is upregulated in urine from MMTV-PyMT mice, was suppressed in mice receiving AAV-mDNase I compared to an AAV-null control group. Furthermore, the proportion of mice that developed lung metastasis was reduced in the AAV-mDNase I group. Altogether, our data indicate that AAV-mDNase I has the potential to reduce cancer-associated impairment of renal function and development of metastasis. We conclude that AAV-mDNase I could represent a promising therapeutic strategy in metastatic breast cancer.
Collapse
Affiliation(s)
- Melanie Herre
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| | - Kalyani Vemuri
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| | - Jessica Cedervall
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| | - Stefanie Nissl
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| | - Falk Saupe
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| | | | - Henrik Lindman
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Casey A. Maguire
- Department of NeurologyHarvard Medical School, Massachusetts General HospitalBostonMassachusettsUSA
- Molecular Neurogenetics UnitMassachusetts General HospitalCharlestownMassachusettsUSA
| | - George Tetz
- CLS TherapeuticsNew YorkNew YorkUSA
- Human Microbiology InstituteDepartment of Systems BiologyNew YorkNew YorkUSA
| | - Victor Tetz
- Human Microbiology InstituteDepartment of Systems BiologyNew YorkNew YorkUSA
| | - Anna‐Karin Olsson
- Department of Medical Biochemistry and Microbiology, Biomedical CenterUppsala UniversityUppsalaSweden
| |
Collapse
|
47
|
Ma Y, Zhao Y, Zhang X. Factors affecting neutrophil functions during sepsis: human microbiome and epigenetics. J Leukoc Biol 2024; 116:672-688. [PMID: 38734968 DOI: 10.1093/jleuko/qiae107] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a severe disease that occurs when the body's immune system reacts excessively to infection. The body's response, which includes an intense antibacterial reaction, can damage its tissues and organs. Neutrophils are the major components of white blood cells in circulation, play a vital role in innate immunity while fighting against infections, and are considered a feature determining sepsis classification. There is a plethora of basic research detailing neutrophil functioning, among which, the study of neutrophil extracellular traps is providing novel insights into mechanisms and treatments of sepsis. This review explores their functions, dysfunctions, and influences in the context of sepsis. The interplay between neutrophils and the human microbiome and the impact of DNA methylation on neutrophil function in sepsis are crucial areas of study. The interaction between neutrophils and the human microbiome is complex, particularly in the context of sepsis, where dysbiosis may occur. We highlight the importance of deciphering neutrophils' functional alterations and their epigenetic features in sepsis because it is critical for defining sepsis endotypes and opening up the possibility for novel diagnostic methods and therapy. Specifically, epigenetic signatures are pivotal since they will provide a novel implication for a sepsis diagnostic method when used in combination with the cell-free DNA. Research is exploring how specific patterns of DNA methylation in neutrophils, detectable in cell-free DNA, could serve as biomarkers for the early detection of sepsis.
Collapse
Affiliation(s)
- Yina Ma
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Xin Zhang
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| |
Collapse
|
48
|
Geng X, Wang DW, Li H. The pivotal role of neutrophil extracellular traps in cardiovascular diseases: Mechanisms and therapeutic implications. Biomed Pharmacother 2024; 179:117289. [PMID: 39151311 DOI: 10.1016/j.biopha.2024.117289] [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: 05/30/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
Cardiovascular diseases (CVDs) continue to pose a significant burden on global health, prominently contributing to morbidity and mortality rates worldwide. Recent years have witnessed an increasing recognition of the intricate involvement of neutrophil extracellular traps (NETs) in the pathology of diverse cardiovascular conditions. This review provides a comprehensive analysis of the multifaceted functions of NETs in cardiovascular diseases, shedding light on the impact on atherosclerosis, myocardial infarction, heart failure, myocarditis, atrial fibrillation, aortic stenosis, and the potential therapeutic avenues targeting NETs.
Collapse
Affiliation(s)
- Xinyu Geng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huihui Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
49
|
Maldonado F, Albornoz M, Enríquez I, Espinoza C, Chang H, Carrasco L, Díaz-Papapietro C, Medina F, González R, Cáceres M. Association of neutrophil-to-lymphocyte ratio with age and 180-day mortality after emergency surgery. BMC Anesthesiol 2024; 24:329. [PMID: 39289610 PMCID: PMC11406743 DOI: 10.1186/s12871-024-02718-z] [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: 02/26/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND To examine the relationship between neutrophil-to-lymphocyte ratio (NLR), age, and mortality rates after emergency surgery. METHODS In this observational study, a total of 851 patients undergoing emergency surgery between January 2022 and January 2023 were retrospective examined. Using 30 and 180 days mortality data, NLR differences and receiver operating characteristic (ROC) curves were analyzed using a 65-year threshold. A multiple logistic regression model was constructed incorporating age and NLR. Finally, Kaplan-Meier curves were constructed for mortality. RESULTS Among 851 patients, the 30 and 180 days mortality rates were 5.2% and 10.8%, respectively. Median NLR in 30 days was 5.6 (3.1 to 9.6) in survivors and 8.7 (4.6 to 13.4) in deceased patients (p < 0.0001); in 180 days, it was 5.5 (3.1 to 9.8) and 8.8 (4.8 to 14.5), respectively (p < 0.0001). In the 30- and 180-days mortality analyses, median NLRs were 5.1 (2.9 to 8.9) and 4.9 (2.9 to 8.8) in survivors and 10.6 (6.9 to 16.6) and 9.3 (5.4 to 14.9) in deceased patients aged < 65 years, respectively. The ROC AUC in patients younger than 65 years was higher for 30 days (AUC 0.75; 95% CI 0.72 to 0.87) and 180 days (AUC 0.73; 95% CI 0.64 to 0.81). Multivariate logistic regression revealed that the NLR (odds ratio, 1.03 [95% CI 1.005 to 1.053; p = 0.0133) and age (odds ratio, 1.05 [95% CI 1.034 to 1.064; p < 0.0001) significantly contributed to the model. Survival analysis revealed differences in the 180 days mortality (p = 0.0006). CONCLUSION We observed differences in preoperative NLR between patients who survived and those who died after emergency surgery. Age impacts the use of NLR as a mortality risk factor. TRIAL REGISTRATION NCT06549101, retrospectively registered.
Collapse
Affiliation(s)
- Felipe Maldonado
- Department of Anesthesia and Perioperative Medicine, Faculty of Medicine, Hospital Clínico de la Universidad de Chile, Universidad de Chile, Santiago, Chile.
| | | | | | | | - Hui Chang
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Laura Carrasco
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Catalina Díaz-Papapietro
- Department of Anesthesia and Perioperative Medicine, Faculty of Medicine, Hospital Clínico de la Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Felipe Medina
- Instituto de Salud Poblacional, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Roberto González
- Department of Anesthesia and Perioperative Medicine, Faculty of Medicine, Hospital Clínico de la Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Mónica Cáceres
- Cellular and Molecular Biology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
50
|
Yao W, Hu X, Wang X. Crossing epigenetic frontiers: the intersection of novel histone modifications and diseases. Signal Transduct Target Ther 2024; 9:232. [PMID: 39278916 PMCID: PMC11403012 DOI: 10.1038/s41392-024-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/30/2024] [Indexed: 09/18/2024] Open
Abstract
Histone post-translational modifications (HPTMs), as one of the core mechanisms of epigenetic regulation, are garnering increasing attention due to their close association with the onset and progression of diseases and their potential as targeted therapeutic agents. Advances in high-throughput molecular tools and the abundance of bioinformatics data have led to the discovery of novel HPTMs which similarly affect gene expression, metabolism, and chromatin structure. Furthermore, a growing body of research has demonstrated that novel histone modifications also play crucial roles in the development and progression of various diseases, including various cancers, cardiovascular diseases, infectious diseases, psychiatric disorders, and reproductive system diseases. This review defines nine novel histone modifications: lactylation, citrullination, crotonylation, succinylation, SUMOylation, propionylation, butyrylation, 2-hydroxyisobutyrylation, and 2-hydroxybutyrylation. It comprehensively introduces the modification processes of these nine novel HPTMs, their roles in transcription, replication, DNA repair and recombination, metabolism, and chromatin structure, as well as their involvement in promoting the occurrence and development of various diseases and their clinical applications as therapeutic targets and potential biomarkers. Moreover, this review provides a detailed overview of novel HPTM inhibitors targeting various targets and their emerging strategies in the treatment of multiple diseases while offering insights into their future development prospects and challenges. Additionally, we briefly introduce novel epigenetic research techniques and their applications in the field of novel HPTM research.
Collapse
Affiliation(s)
- Weiyi Yao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|