1
|
Sun Y, He J, Chen W, Wang Y, Wang K, Zhou M, Zheng Y. Inhalable DNase I@Au hybrid nanoparticles for radiation sensitization and metastasis inhibition by elimination of neutrophil extracellular traps. Biomaterials 2025; 317:123095. [PMID: 39813970 DOI: 10.1016/j.biomaterials.2025.123095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
High-dose radiation therapy is a widely used clinical strategy to inhibit tumor growth. However, the rapid generation of excessive reactive oxygen species (ROS) triggers the formation of neutrophil extracellular traps (NETs), which capture free tumor cells in the bloodstream, promoting metastasis. In this study, we developed a hybrid nanoparticle composed of DNase I and gold (DNase I@Au) to enhance radiotherapy efficacy while mitigating metastasis by precisely eliminating NETs. The DNase I@Au nanoparticles, administered via aerosol inhalation, are efficiently delivered to lung tumor tissue, improving radiosensitization and reducing tumor size. Crucially, the nanoparticles could gradually release DNase I, effectively degrading ROS-induced NETs and preventing the interaction of free malignant cells with tumor sites or vasculature, thereby inhibiting metastasis. Therefore, we provide an enzyme and sensitizer co-loaded strategy that offers a promising approach to improve the therapeutic outcome of radiotherapy and reduce the risk of lung cancer metastasis under ROS stimulation.
Collapse
Affiliation(s)
- Yuchao Sun
- Department of Urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jian He
- University-University of Edinburgh Institute (ZJU-UoE Institute), and liangzhu Laboratory, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Weiyu Chen
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Yongfang Wang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Min Zhou
- Department of Urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; University-University of Edinburgh Institute (ZJU-UoE Institute), and liangzhu Laboratory, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China; Key Laboratory of Cancer Prevention and Intervention of China (MOE), Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Yichun Zheng
- Department of Urology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Xu Y, Ren Y, Zou W, Wang M, Ye X, Shen W. Neutrophil extracellular traps aggravate uremic cardiomyopathy by inducing myocardial fibroblast pyroptosis. Sci Rep 2025; 15:17509. [PMID: 40394287 PMCID: PMC12092652 DOI: 10.1038/s41598-025-02383-3] [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: 12/05/2024] [Accepted: 05/13/2025] [Indexed: 05/22/2025] Open
Abstract
Uremic cardiomyopathy (UCM) is a leading cause of death in patients with chronic kidney disease. This study aimed to explore the role and underlying mechanisms of neutrophil extracellular traps (NETs) in UCM. A UCM mouse model was constructed using 5/6 nephrectomy and treated with the NET inhibitor DNaseI. Cardiac fibroblasts were incubated with 200 ng/mL of NETs to establish an in vitro model. Physiological parameters of mice were measured, and commercial kits were used to detect MPO-DNA, IL-1β, and IL-6 levels. Hematoxylin-eosin and TUNEL staining were performed to evaluate myocardial injury and apoptosis. Immunofluorescence was used to detect MPO, CitH3, and caspase-1 expression. Sytox Green staining and CCK-8 assay were applied to assess NET formation and determine the optimal NET concentration. Western blot was conducted to detect collagen1, α-SMA, NLRP3, GSDMD-N, and caspase-1 expression. Transmission electron microscopy was performed to observe pyroptosis. UCM mice exhibited impaired kidney and cardiac function, indicating the successful establishment of the UCM model. MPO-DNA levels were elevated in UCM mice, suggesting NET formation. DNaseI treatment significantly reduced MPO and CitH3 expression, inhibiting NET formation in UCM mice. NET inhibition improved renal function and alleviated cardiac injury. Furthermore, NET inhibition reduced inflammation, apoptosis, and pyroptosis in UCM mice. Neutrophils isolated from UCM mice, as well as PMA, induced NET formation. NETs significantly enhanced fibrosis and pyroptosis in myocardial fibroblasts. NETs may promote UCM progression by inducing pyroptosis in myocardial fibroblasts.
Collapse
Affiliation(s)
- Ying Xu
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Ren
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wenli Zou
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Minmin Wang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xianyun Ye
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wei Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
3
|
Yaron JR, Pallod S, Grigaitis-Esman N, Singh V, Rhodes S, Patel DM, Ghosh D, Rege K. Histamine receptor agonism differentially induces immune and reparative healing responses in biomaterial-facilitated tissue repair. Biomaterials 2025; 315:122967. [PMID: 39586217 DOI: 10.1016/j.biomaterials.2024.122967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
Tissue repair is a highly regulated process involving immune, stromal, vascular, and parenchymal cell responses. Mediators of cellular responses at different phases of the healing process stimulate transitions through the continuum of repair. Histamine is an early mediator of healing, which, in skin, is released by resident cells (e.g., mast cells) after cutaneous injury, and acts to stimulate diverse responses in multiple cell populations. Histamine signaling is regulated by four distinct cell surface G-protein coupled receptors (HRH1-4 in humans, Hrh1-4 in mice) which initiate different downstream signaling cascades upon activation, but the specific effect of each receptor on tissue repair is poorly understood. Here, we systematically investigated the effect of selective histamine receptor agonism in laser-activated sealing and tissue repair of incisional skin wounds in immunocompetent mice. Although all four histamine receptors exhibited wound responsiveness in the epidermis, we find that activation of Hrh1, Hrh2, and Hrh4 stimulate a pro-healing immune response characterized by increased pro-resolution macrophages, reduced pro-inflammatory macrophages, and suppressed neutrophil responses. Further, activation of Hrh1 and Hrh4 stimulate angiogenesis after injury. Lastly, although Hrh1 activation resulted in enhanced epidermal epithelial-to-mesenchymal transition (EMT) in vivo and epithelialization in vitro, activation of Hrh2 suppressed both epidermal EMT and epithelialization. Activation of Hrh3, primarily found on neuronal cells, had no effect on any measure in our study. Selective histamine receptor agonism, specifically of histamine receptors Hrh-1 and 4, is a potential reparative approach to promote the efficacy of biomaterial-mediated repair of tissues, including skin.
Collapse
Affiliation(s)
- Jordan R Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Shubham Pallod
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Nicole Grigaitis-Esman
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Vanshika Singh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Samantha Rhodes
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Dirghau Manishbhai Patel
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Deepanjan Ghosh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
4
|
Manoj H, Gomes SM, Thimmappa PY, Nagareddy PR, Jamora C, Joshi MB. Cytokine signalling in formation of neutrophil extracellular traps: Implications for health and diseases. Cytokine Growth Factor Rev 2025; 81:27-39. [PMID: 39681501 DOI: 10.1016/j.cytogfr.2024.12.001] [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: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Neutrophils, as essential component of the innate immune response, form a crucial part in the defence mechanisms through the release of extracellular traps (NETs). These web-like structures, composed of chromatin and antimicrobial proteins, are essential for the entrapment and inactivation of pathogens. However, either constitutive formation or inefficient clearance of NETs leads to adverse effects such as fibrosis, thrombosis, delayed wound healing and tissue damage in multiple diseases associated with sterile inflammation. This dichotomy casts NETs as both protective agents and harmful factors in several diseases such as autoimmune diseases, metabolic syndromes, systemic infections, and malignancies. Besides microbes and their products, variety of stimulants including pro-inflammatory cytokines induce NETs. The complex interactions and cross talk among the pro-inflammatory cytokines including IL-8, IL-6, GM-CSF, TNF-α, IFNs, and IL-1β activate neutrophils to form NETs and also contributes to a vicious circle of inflammatory cascade, leading to increased inflammation, oxidative stress, and thrombotic events. Emerging evidence indicates that the dysregulated cytokine milieus in diseases, such as diabetes mellitus, obesity, atherosclerosis, stroke, rheumatoid arthritis, and systemic lupus erythematosus, potentiate NETs release, thereby promoting disease development. Thus, neutrophils represent both critical effectors and potential therapeutic targets, underscoring their importance in the context of cytokine-mediated therapies for a spectrum of diseases. In the present review, we describe various cytokines and associated signalling pathways activating NETs formation in different human pathologies. Further, the review identifies potential strategies to pharmacologically modulate cytokine pathways to reduce NETs.
Collapse
Affiliation(s)
- Haritha Manoj
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sarah Michael Gomes
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma, OK, USA
| | - Colin Jamora
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh 201314, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
5
|
Xiong T, Qin W, Zhang Y, Chen Y, Ou Y. Prognostic nutritional index and naples prognostic score as biomarkers for the prognosis of incisional wound healing after thoracolumbar tuberculosis surgery. PLoS One 2024; 19:e0309267. [PMID: 39671379 PMCID: PMC11642951 DOI: 10.1371/journal.pone.0309267] [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: 10/14/2023] [Accepted: 08/07/2024] [Indexed: 12/15/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate and compare the clinical predictive value of prognostic nutritional index (PNI) and naples prognostic score (NPS) as biomarkers for the prognosis of incisional wound healing in patients who underwent thoracolumbar tuberculosis surgery through the posterior approach. METHODS From January 2019 to October 2021, a total of 124 patients with thoracolumbar tuberculosis who underwent posterior approach debridement and internal fixation were included in this study. We retrospectively analyzed the clinical data, including PNI and NPS. They were divided into poor wound healing (PWH) and non-PWH groups according to whether PWH developed after the operation. And according to the receiver operating characteristic curve, patients were divided into two groups through the threshold value. Risk factors were found using logistic regression analysis. RESULTS The unfavorable outcome group had lower hemoglobin, serum albumin, Pre-albumin, PNI, and higher estimated blood loss, instrumented segments, neutrophil count, and NPS (P < 0.05). Both PNI and NPS were strongly correlated with PWH (r = 0.373, P < 0.05; r = -0.306, P < 0.05, respectively). The area under the curve (AUC) of PNI for predicting unfavorable outcomes was 0.764 (95% CI 0.662-0.865, P < 0.001), which was similar to NPS (0.808, 95% CI: 0.719-0.897, P < 0.001). Multivariate stepwise logistic regression analysis showed that PNI, NPS, the neutrophil count, the level of serum albumin, and the number of instrumented segments were independent risk factors for PWH. CONCLUSION Both PNI and NPS might be novel independent biomarkers and predictors of poor outcomes in incisional wound healing after STB surgery.
Collapse
Affiliation(s)
- Tuotuo Xiong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
- Department of Orthopaedic Surgery, Chongqing university Jiangjin hospital, Chongqing, China
| | - Wanyuan Qin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Ye Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Yuxing Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Yunsheng Ou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Zhu Z, Zhou S, Li S, Gong S, Zhang Q. Neutrophil extracellular traps in wound healing. Trends Pharmacol Sci 2024; 45:1033-1045. [PMID: 39419742 DOI: 10.1016/j.tips.2024.09.007] [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: 07/19/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
Wound healing is a complex and orchestrated process that involves hemostasis, inflammation, proliferation, and tissue remodeling. Neutrophil extracellular traps (NETs) are intricate web-like structures released by neutrophils, comprising decondensed chromatin, myeloperoxidase (MPO), and neutrophil elastase (NE), which play vital roles in regulating neutrophil-mediated immune regulation. While NETs contribute to wound healing, excessive activation induced by dysregulated inflammation can hinder the healing process. Understanding the pivotal role of NETs in wound healing and tissue remodeling, as well as their intricate interactions within the wound microenvironment, presents opportunities for innovative wound healing strategies. In this review we discuss the process of NET formation, explore the interactions between NETs and skin cells, and examine therapeutic strategies targeting NETs and drug delivery platforms to accelerate wound healing. Additionally, we discuss current clinical investigations and research challenges towards advancing wound care practices.
Collapse
Affiliation(s)
- Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Shengzhi Zhou
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan 430030, Hubei Province, People's Republic of China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China.
| |
Collapse
|
7
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
8
|
Lu Y, Elrod J, Herrmann M, Knopf J, Boettcher M. Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation. Cells 2024; 13:991. [PMID: 38891123 PMCID: PMC11171752 DOI: 10.3390/cells13110991] [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: 04/07/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Post-surgical abdominal adhesions, although poorly understood, are highly prevalent. The molecular processes underlying their formation remain elusive. This review aims to assess the relationship between neutrophil extracellular traps (NETs) and the generation of postoperative peritoneal adhesions and to discuss methods for mitigating peritoneal adhesions. A keyword or medical subject heading (MeSH) search for all original articles and reviews was performed in PubMed and Google Scholar. It included studies assessing peritoneal adhesion reformation after abdominal surgery from 2003 to 2023. After assessing for eligibility, the selected articles were evaluated using the Critical Appraisal Skills Programme checklist for qualitative research. The search yielded 127 full-text articles for assessment of eligibility, of which 7 studies met our criteria and were subjected to a detailed quality review using the Critical Appraisal Skills Programme (CASP) checklist. The selected studies offer a comprehensive analysis of adhesion pathogenesis with a special focus on the role of neutrophil extracellular traps (NETs) in the development of peritoneal adhesions. Current interventional strategies are examined, including the use of mechanical barriers, advances in regenerative medicine, and targeted molecular therapies. In particular, this review emphasizes the potential of NET-targeted interventions as promising strategies to mitigate postoperative adhesion development. Evidence suggests that in addition to their role in innate defense against infections and autoimmune diseases, NETs also play a crucial role in the formation of peritoneal adhesions after surgery. Therefore, therapeutic strategies that target NETs are emerging as significant considerations for researchers. Continued research is vital to fully elucidate the relationship between NETs and post-surgical adhesion formation to develop effective treatments.
Collapse
Affiliation(s)
- Yuqing Lu
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jasmin Knopf
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
9
|
Zang T, Fear MW, Parker TJ, Holland AJA, Martin L, Langley D, Kimble R, Wood FM, Cuttle L. Inflammatory proteins and neutrophil extracellular traps increase in burn blister fluid 24h after burn. Burns 2024; 50:1180-1191. [PMID: 38490838 DOI: 10.1016/j.burns.2024.02.026] [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: 05/16/2023] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024]
Abstract
Burn wound blister fluid is a valuable matrix for understanding the biological pathways associated with burn injury. In this study, 152 blister fluid samples collected from paediatric burn wounds at three different hospitals were analysed using mass spectrometry proteomic techniques. The protein abundance profile at different days after burn indicated more proteins were associated with cellular damage/repair in the first 24 h, whereas after this point more proteins were associated with antimicrobial defence. The inflammatory proteins persisted at a high level in the blister fluid for more than 7 days. This may indicate that removal of burn blisters prior to two days after burn is optimal to prevent excessive or prolonged inflammation in the wound environment. Additionally, many proteins associated with the neutrophil extracellular trap (NET) pathway were increased after burn, further implicating NETs in the post-burn inflammatory response. NET inhibitors may therefore be a potential treatment to reduce post-burn inflammation and coagulation pathology and enhance burn wound healing outcomes.
Collapse
Affiliation(s)
- Tuo Zang
- Queensland University of Technology (QUT), Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, South Brisbane, Queensland, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Tony J Parker
- Queensland University of Technology (QUT), School of Biomedical Sciences, Faculty of Health, Kelvin Grove, Queensland, Australia
| | - Andrew J A Holland
- The Children's Hospital at Westmead Burns Unit, Kids Research Institute, Department of Paediatrics and Child Health, Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Lisa Martin
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Donna Langley
- Queensland University of Technology (QUT), Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, South Brisbane, Queensland, Australia
| | - Roy Kimble
- Children's Health Queensland, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital and Fiona Stanley Hospital, Perth, WA, Australia
| | - Leila Cuttle
- Queensland University of Technology (QUT), Faculty of Health, School of Biomedical Sciences, Centre for Children's Health Research, South Brisbane, Queensland, Australia.
| |
Collapse
|
10
|
Ma H, Yao W, Peng B, Liu X, Chen J, Lin Y, Di T, Li P, He X. Mercury-containing preparations attenuate neutrophil extracellular trap formation in mice and humans through inhibiting the ERK1/2 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117421. [PMID: 37979820 DOI: 10.1016/j.jep.2023.117421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/30/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neutrophil extracellular trap (NET) formation plays a crucial role in wound healing disorders, including chronic skin ulcers and diabetic foot ulcers (DFUs). Over the years, traditional Chinese topical medications, such as Cinnabar (composed of HgS and soluble mercury salt) and hydrargyria oxydum rubrum (containing HgO and soluble mercury salt), have been utilized for treating these ailments. Nevertheless, the fundamental processes remain mostly ambiguous. AIM OF THE STUDY This study sought to investigate the potential effects of topical mercury-containing preparations on the process of NET formation. MATERIALS AND METHODS Neutrophils isolated from healthy individuals and mouse models of type 1 and type 2 diabetes were cultured with phorbol 12-myristate 13-acetate (PMA), both with and without the mercury-containing preparations (MCP). The formation of NETs was monitored using confocal and scanning electron microscopes. Immunofluorescence and fluorescent probes were employed to assess the levels of citrulline histone H3 (Cit-H3) and intracellular reactive oxygen species (ROS), respectively. The impact of MCP extracts on cytokine expression, peptidylarginine deiminase 4 (PAD4), and myeloperoxidase (MPO) was measured through Luminex and ELISA assays. Phagocytosis of human neutrophils was analyzed using Flow Cytometry. Finally, the phosphorylation levels of ERK were detected by western blotting. RESULTS Treatment with MCP led to a reduction in PAD4, Cit-H3, and MPO expressions in neutrophils, consequently inhibiting PMA-induced NET formation. MCP treatment also dampened ERK1/2 activation in neutrophils. Furthermore, MCP exhibited inhibitory effects on the secretion of the cytokine IL-8 and ROS production while enhancing neutrophil phagocytosis. CONCLUSION Our findings suggest that MCP can mitigate the release of NETs, likely by suppressing the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Wentao Yao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Bing Peng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Xin Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Jia Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Yan Lin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China.
| | - Xiujuan He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing, 100010, China.
| |
Collapse
|
11
|
Yang R, Zhang Y, Kang J, Zhang C, Ning B. Chondroitin Sulfate Proteoglycans Revisited: Its Mechanism of Generation and Action for Spinal Cord Injury. Aging Dis 2024; 15:153-168. [PMID: 37307832 PMCID: PMC10796098 DOI: 10.14336/ad.2023.0512] [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: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 06/14/2023] Open
Abstract
Reactive astrocytes (RAs) produce chondroitin sulfate proteoglycans (CSPGs) in large quantities after spinal cord injury (SCI) and inhibit axon regeneration through the Rho-associated protein kinase (ROCK) pathway. However, the mechanism of producing CSPGs by RAs and their roles in other aspects are often overlooked. In recent years, novel generation mechanisms and functions of CSPGs have gradually emerged. Extracellular traps (ETs), a new recently discovered phenomenon in SCI, can promote secondary injury. ETs are released by neutrophils and microglia, which activate astrocytes to produce CSPGs after SCI. CSPGs inhibit axon regeneration and play an important role in regulating inflammation as well as cell migration and differentiation; some of these regulations are beneficial. The current review summarized the process of ET-activated RAs to generate CSPGs at the cellular signaling pathway level. Moreover, the roles of CSPGs in inhibiting axon regeneration, regulating inflammation, and regulating cell migration and differentiation were discussed. Finally, based on the above process, novel potential therapeutic targets were proposed to eliminate the adverse effects of CSPGs.
Collapse
Affiliation(s)
- Rui Yang
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianning Kang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ce Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
12
|
Willemsen JF, Wenskus J, Lenz M, Rhode H, Trochimiuk M, Appl B, Pagarol-Raluy L, Börnigen D, Bang C, Reinshagen K, Herrmann M, Elrod J, Boettcher M. DNases improve effectiveness of antibiotic treatment in murine polymicrobial sepsis. Front Immunol 2024; 14:1254838. [PMID: 38259485 PMCID: PMC10801052 DOI: 10.3389/fimmu.2023.1254838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/07/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Neutrophil extracellular traps (NETs) have various beneficial and detrimental effects in the body. It has been reported that some bacteria may evade the immune system when entangled in NETs. Thus, the aim of the current study was to evaluate the effects of a combined DNase and antibiotic therapy in a murine model of abdominal sepsis. Methods C57BL/6 mice underwent a cecum-ligation-and-puncture procedure. We used wild-type and knockout mice with the same genetic background (PAD4-KO and DNase1-KO). Mice were treated with (I) antibiotics (Metronidazol/Cefuroxime), (II) DNAse1, or (III) with the combination of both; mock-treated mice served as controls. We employed a streak plate procedure and 16s-RNA analysis to evaluate bacterial translocation and quantified NETs formation by ELISA and immune fluorescence. Western blot and proteomics analysis were used to determine inflammation. Results A total of n=73 mice were used. Mice that were genetically unable to produce extended NETs or were treated with DNases displayed superior survival and bacterial clearance and reduced inflammation. DNase1 treatment significantly improved clearance of Gram-negative bacteria and survival rates. Importantly, the combination of DNase1 and antibiotics reduced tissue damage, neutrophil activation, and NETs formation in the affected intestinal tissue. Conclusion The combination of antibiotics with DNase1 ameliorates abdominal sepsis. Gram-negative bacteria are cleared better when NETs are cleaved by DNase1. Future studies on antibiotic therapy should be combined with anti-NETs therapies.
Collapse
Affiliation(s)
- Jan-Fritjof Willemsen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Wenskus
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Moritz Lenz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rhode
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madgalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laia Pagarol-Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniela Börnigen
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
- Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitäts-klinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Elrod
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
13
|
Elrod J, Heuer A, Knopf J, Schoen J, Schönfeld L, Trochimiuk M, Stiel C, Appl B, Raluy LP, Saygi C, Zlatar L, Hosari S, Royzman D, Winkler TH, Lochnit G, Leppkes M, Grützmann R, Schett G, Tomuschat C, Reinshagen K, Herrmann M, Fuchs TA, Boettcher M. Neutrophil extracellular traps and DNases orchestrate formation of peritoneal adhesions. iScience 2023; 26:108289. [PMID: 38034352 PMCID: PMC10682263 DOI: 10.1016/j.isci.2023.108289] [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: 01/17/2023] [Revised: 08/05/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023] Open
Abstract
Peritoneal adhesions are poorly understood but highly prevalent conditions that can cause intestinal obstruction and pelvic pain requiring surgery. While there is consensus that stress-induced inflammation triggers peritoneal adhesions, the molecular processes of their formation still remain elusive. We performed murine models and analyzed human samples to monitor the formation of adhesions and the treatment with DNases. Various molecular analyses were used to evaluate the adhesions. The experimental peritoneal adhesions of the murine models and biopsy material from humans are largely based on neutrophil extracellular traps (NETs). Treatment with DNASE1 (Dornase alfa) and the human DNASE1L3 analog (NTR-10), significantly reduced peritoneal adhesions in experimental models. We conclude that NETs serve as essential scaffold for the formation of adhesions; DNases interfere with this process. Herein, we show that therapeutic application of DNases can be employed to prevent the formation of murine peritoneal adhesions. If this can be translated into the human situation requires clinical studies.
Collapse
Affiliation(s)
- Julia Elrod
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Annika Heuer
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Knopf
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina Schoen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lavinia Schönfeld
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carolin Stiel
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ceren Saygi
- Bioinformatics Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leticija Zlatar
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sami Hosari
- Department of Surgery, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dmytro Royzman
- Division of Genetics, Department of Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department of Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Faculty of Medicine, Justus Liebig University Giessen, Friedrichstrasse 24, Giessen, Germany
| | - Moritz Leppkes
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1 - Gastroenterologie, Pneumologie und Endokrinologie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias A. Fuchs
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
14
|
Liu Y, Ma YH, Yang JW, Man JW, Wang HB, Li Y, Liang C, Cao JL, Chen SY, Li KP, Yang L. Rethinking neutrophil extracellular traps. Int Immunopharmacol 2023; 124:110834. [PMID: 37625368 DOI: 10.1016/j.intimp.2023.110834] [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: 05/17/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Neutrophils are a major subset of leukocytes in human circulating blood. In some circumstances, neutrophils release neutrophil extracellular traps (NETs). lnitially, NETs were considered to have a strong antibacterial capacity. However, currently, NETs have been shown to have a pivotal impact on various diseases. Different stimulators induce the production of different types of NETs, and their biological functions and modes of clearance do not appear to be the same. In this review, we will discuss several important issues related to NETs in order to better understand the relationship between NETs and diseases, as well as how to utilize the characteristics of NETs for disease treatment.
Collapse
Affiliation(s)
- Yi Liu
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Yu-Hua Ma
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jian-Wei Yang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jiang-Wei Man
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Hua-Bin Wang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Yi Li
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Cheng Liang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jin-Long Cao
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China.
| |
Collapse
|
15
|
Linnemann C, Sahin F, Chen Y, Falldorf K, Ronniger M, Histing T, Nussler AK, Ehnert S. NET Formation Was Reduced via Exposure to Extremely Low-Frequency Pulsed Electromagnetic Fields. Int J Mol Sci 2023; 24:14629. [PMID: 37834077 PMCID: PMC10572227 DOI: 10.3390/ijms241914629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Fracture-healing is a highly complex and timely orchestrated process. Non-healing fractures are still a major clinical problem and treatment remains difficult. A 16 Hz extremely low-frequency pulsed electromagnetic field (ELF-PEMF) was identified as non-invasive adjunct therapy supporting bone-healing by inducing reactive oxygen species (ROS) and Ca2+-influx. However, ROS and Ca2+-influx may stimulate neutrophils, the first cells arriving at the wounded site, to excessively form neutrophil extracellular traps (NETs), which negatively affects the healing process. Thus, this study aimed to evaluate the effect of this 16 Hz ELF-PEMF on NET formation. Neutrophils were isolated from healthy volunteers and exposed to different NET-stimuli and the 16 Hz ELF-PEMF. NETs were quantified using Sytox Green Assay and immunofluorescence, Ca2+-influx and ROS with fluorescence probes. In contrast to mesenchymal cells, ELF-PEMF exposure did not induce ROS and Ca2+-influx in neutrophils. ELF-PEMF exposure did not result in basal or enhanced PMA-induced NET formation but did reduce the amount of DNA released. Similarly, NET formation induced by LPS and H2O2 was reduced through exposure to ELF-PEMF. As ELF-PEMF exposure did not induce NET release or negatively affect neutrophils, the ELF-PEMF exposure can be started immediately after fracture treatment.
Collapse
Affiliation(s)
- Caren Linnemann
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Filiz Sahin
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Yangmengfan Chen
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Michael Ronniger
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Tina Histing
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| |
Collapse
|
16
|
Moon S, Hong J, Go S, Kim BS. Immunomodulation for Tissue Repair and Regeneration. Tissue Eng Regen Med 2023; 20:389-409. [PMID: 36920675 PMCID: PMC10219918 DOI: 10.1007/s13770-023-00525-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
17
|
Singh J, Boettcher M, Dölling M, Heuer A, Hohberger B, Leppkes M, Naschberger E, Schapher M, Schauer C, Schoen J, Stürzl M, Vitkov L, Wang H, Zlatar L, Schett GA, Pisetsky DS, Liu ML, Herrmann M, Knopf J. Moonlighting chromatin: when DNA escapes nuclear control. Cell Death Differ 2023; 30:861-875. [PMID: 36755071 PMCID: PMC9907214 DOI: 10.1038/s41418-023-01124-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 02/10/2023] Open
Abstract
Extracellular chromatin, for example in the form of neutrophil extracellular traps (NETs), is an important element that propels the pathological progression of a plethora of diseases. DNA drives the interferon system, serves as autoantigen, and forms the extracellular scaffold for proteins of the innate immune system. An insufficient clearance of extruded chromatin after the release of DNA from the nucleus into the extracellular milieu can perform a secret task of moonlighting in immune-inflammatory and occlusive disorders. Here, we discuss (I) the cellular events involved in the extracellular release of chromatin and NET formation, (II) the devastating consequence of a dysregulated NET formation, and (III) the imbalance between NET formation and clearance. We include the role of NET formation in the occlusion of vessels and ducts, in lung disease, in autoimmune diseases, in chronic oral disorders, in cancer, in the formation of adhesions, and in traumatic spinal cord injury. To develop effective therapies, it is of utmost importance to target pathways that cause decondensation of chromatin during exaggerated NET formation and aggregation. Alternatively, therapies that support the clearance of extracellular chromatin are conceivable.
Collapse
Affiliation(s)
- Jeeshan Singh
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maximilian Dölling
- Department of Surgery, University Hospital Magdeburg, Magdeburg, Germany
| | - Annika Heuer
- Division of Spine Surgery, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Mildred-Scheel Cancer Career Center Hamburg HaTriCS4, University Cancer Center Hamburg, Hamburg, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Moritz Leppkes
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Gastroenterology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universtität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mirco Schapher
- Department of Otorhinolaryngology, Head and Neck Surgery, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus University, Nürnberg, Germany
| | - Christine Schauer
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina Schoen
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universtität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ljubomir Vitkov
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, Homburg, Germany
- Department of Environment & Biodiversity, University of Salzburg, Salzburg, 5020, Austria
- Department of Dental Pathology, University of East Sarajevo, East Sarajevo, Republic of Srpska, Bosnia and Herzegovina
| | - Han Wang
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Leticija Zlatar
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg A Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David S Pisetsky
- Department of Medicine and Immunology and Medical Research Service, Duke University Medical Center and Veterans Administration Medical Center, Durham, NC, USA
| | - Ming-Lin Liu
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA
| | - Martin Herrmann
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| | - Jasmin Knopf
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
18
|
Klinke M, Chaaban H, Boettcher M. The role of neutrophil extracellular traps in necrotizing enterocolitis. Front Pediatr 2023; 11:1121193. [PMID: 37009300 PMCID: PMC10050739 DOI: 10.3389/fped.2023.1121193] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/27/2023] [Indexed: 04/04/2023] Open
Abstract
Necrotizing enterocolitis (NEC) continues to be one of the most common causes of mortality and morbidity in preterm infants. Although not fully elucidated, studies suggest that prematurity, formula feeding, imbalanced vascular supply, and altered bacterial colonization play major roles in the pathogenesis of NEC. NEC is characterized by increased cytokine release and leukocyte infiltration. Recent data from preterm infants and animal models of NEC suggest that neutrophil extracellular traps (NETs) are released in intestinal tissue. The contribution of NETs in the pathogenesis and/or prevention/treatment of this disease continues to be controversial. Here, we review the available data on NETs release in NEC in human patients and in different NEC models, highlighting their potential contribution to pathology and resolution of inflammation. Here, we review the available data on NETs release in NEC in human patients and the different NEC models, highlighting their potential contribution to pathology or resolution of inflammation.
Collapse
Affiliation(s)
- Michaela Klinke
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hala Chaaban
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
19
|
Ghosh D, Salinas CM, Pallod S, Roberts J, Makin IRS, Yaron JR, Witte RS, Rege K. Temporal evaluation of efficacy and quality of tissue repair upon laser-activated sealing. Bioeng Transl Med 2023; 8:e10412. [PMID: 36925709 PMCID: PMC10013809 DOI: 10.1002/btm2.10412] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022] Open
Abstract
Injuries caused by surgical incisions or traumatic lacerations compromise the structural and functional integrity of skin. Immediate approximation and robust repair of skin are critical to minimize occurrences of dehiscence and infection that can lead to impaired healing and further complication. Light-activated skin sealing has emerged as an alternative to sutures, staples, and superficial adhesives, which do not integrate with tissues and are prone to scarring and infection. Here, we evaluate both shorter- and longer-term efficacy of tissue repair response following laser-activated sealing of full-thickness skin incisions in immunocompetent mice and compare them to the efficacy seen with sutures. Laser-activated sealants (LASEs) in which, indocyanine green was embedded within silk fibroin films, were used to form viscous pastes and applied over wound edges. A hand-held, near-infrared laser was applied over the incision, and conversion of the light energy to heat by the LASE facilitated rapid photothermal sealing of the wound in approximately 1 min. Tissue repair with LASEs was evaluated using functional recovery (transepidermal water loss), biomechanical recovery (tensile strength), tissue visualization (ultrasound [US] and photoacoustic imaging [PAI]), and histology, and compared with that seen in sutures. Our studies indicate that LASEs promoted earlier recovery of barrier and mechanical function of healed skin compared to suture-closed incisions. Visualization of sealed skin using US and PAI indicated integration of the LASE with the tissue. Histological analyses of LASE-sealed skin sections showed reduced neutrophil and increased proresolution macrophages on Days 2 and 7 postclosure of incisions, without an increase in scarring or fibrosis. Together, our studies show that simple fabrication and application methods combined with rapid sealing of wound edges with improved histological outcomes make LASE a promising alternative for management of incisional wounds and lacerations.
Collapse
Affiliation(s)
- Deepanjan Ghosh
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| | | | - Shubham Pallod
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| | - Jordan Roberts
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| | | | - Jordan R. Yaron
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
- Department of Chemical Engineering, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| | - Russell S. Witte
- James C. Wyant College of Optical SciencesUniversity of ArizonaTucsonArizonaUSA
- Department of Medical ImagingUniversity of ArizonaTucsonArizonaUSA
| | - Kaushal Rege
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
- Department of Chemical Engineering, School for Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| |
Collapse
|
20
|
Elrod J, Lenz M, Kiwit A, Armbrust L, Schönfeld L, Reinshagen K, Pagerols Raluy L, Mohr C, Saygi C, Alawi M, Rohde H, Herrmann M, Boettcher M. Murine scald models characterize the role of neutrophils and neutrophil extracellular traps in severe burns. Front Immunol 2023; 14:1113948. [PMID: 36825027 PMCID: PMC9941538 DOI: 10.3389/fimmu.2023.1113948] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Introduction Severe burns cause unique pathophysiological alterations especially on the immune system. A murine scald model was optimized as a basis for the understanding of immunological reactions in response to heat induced injury. The understanding of the roles of neutrophil extracellular traps (NETs) and DNases will support the development of new surgical or pharmacological strategies for the therapy of severe burns. Methods We studied C57BL/6 mice (n=30) and employed four scalding protocols with varying exposure times to hot water. An additional scald group with a shorter observational time was generated to reduce mortality and study the very early phase of pathophysiology. At 24h or 72h, blood was drawn and tissue (wound, liver, lung, spleen) was analyzed for the presence of NETs, oxidative stress, apoptosis, bacterial translocation, and extracellular matrix re-organization. In addition, we analyzed the transcriptome from lung and liver tissues. Results Exposure to hot water for 7s led to significant systemic and local effects and caused considerable late mortality. Therefore, we used an observation time of 24h in this groups. To study later phases of burns (72h) an exposure time of 6s is optimal. Both conditions led to significant disorganization of collagen, increased oxidative stress, NET formation (by immunodetection of H3cit, NE, MPO), apoptosis (cC3) and alterations of the levels of DNase1 and DNase1L3. Transcriptome analysis revealed remarkable alterations in genes involved in acute phase signaling, cell cohesion, extracellular matrix organization, and immune response. Conclusion We identified two scald models that allow the analysis of early (24h) or late (72h) severe burn effects, thereby generating reproducible and standardized scald injuries. The study elucidated the important involvement of neutrophil activity and the role of NETs in burns. Extensive transcriptome analysis characterized the acute phase and tissue remodeling pathways involved in the process of healing and may serve as crucial basis for future in-depth studies.
Collapse
Affiliation(s)
- Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany,Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Julia Elrod,
| | - Moritz Lenz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Kiwit
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lina Armbrust
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lavinia Schönfeld
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Mohr
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Ceren Saygi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany,Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany,Deutsches Zentrum Immuntherapie DZI, Friedrich Alexander University Erlangen-Nuremberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany,Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Ho JW, Quan C, Gauger MA, Alam HB, Li Y. ROLE OF PEPTIDYLARGININE DEIMINASE AND NEUTROPHIL EXTRACELLULAR TRAPS IN INJURIES: FUTURE NOVEL DIAGNOSTICS AND THERAPEUTIC TARGETS. Shock 2023; 59:247-255. [PMID: 36597759 PMCID: PMC9957939 DOI: 10.1097/shk.0000000000002052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
ABSTRACT Injuries lead to an early systemic inflammatory state with innate immune system activation. Neutrophil extracellular traps (NETs) are a complex of chromatin and proteins released from the activated neutrophils. Although initially described as a response to bacterial infections, NETs have also been identified in the sterile postinjury inflammatory state. Peptidylarginine deiminases (PADs) are a group of isoenzymes that catalyze the conversion of arginine to citrulline, termed citrullination or deimination. PAD2 and PAD4 have been demonstrated to play a role in NET formation through citrullinated histone 3. PAD2 and PAD4 have a variety of substrates with variable organ distribution. Preclinical and clinical studies have evaluated the role of PADs and NETs in major trauma, hemorrhage, burns, and traumatic brain injury. Neutrophil extracellular trap formation and PAD activation have been shown to contribute to the postinjury inflammatory state leading to a detrimental effect on organ systems. This review describes our current understanding of the role of PAD and NET formation following injury and burn. This is a new field of study, and the emerging data appear promising for the future development of targeted biomarkers and therapies in trauma.
Collapse
Affiliation(s)
- Jessie W. Ho
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Chao Quan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - Megan A. Gauger
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Hasan B. Alam
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yongqing Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
22
|
Elrod J, Kiwit A, Lenz M, Rohde H, Börnigen D, Alawi M, Mohr C, Pagerols Raluy L, Trochimiuk M, Knopf J, Reinshagen K, Herrmann M, Boettcher M. Midgut Volvulus Adds a Murine, Neutrophil-Driven Model of Septic Condition to the Experimental Toolbox. Cells 2023; 12:cells12030366. [PMID: 36766707 PMCID: PMC9913099 DOI: 10.3390/cells12030366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Severe infections that culminate in sepsis are associated with high morbidity and mortality. Despite continuous efforts in basis science and clinical research, evidence based-therapy is mostly limited to basic causal and supportive measures. Adjuvant therapies often remain without clear evidence. The objective of this study was to evaluate the septic volvulus ischemia-reperfusion model in comparison to two already established models and the role of neutrophil extacellular traps (NETs) in this model. METHODS The technique of the murine model of midgut volvulus was optimized and was compared to two established models of murine sepsis, namely cecal ligation and puncture (CLP) and intra-peritoneal (i.p.) injection of lipopolysaccharide (LPS). RESULTS Midgut volvulus for 15 min caused a comparable mortality (38%) as CLP (55%) and peritoneal LPS injection (25%) at 48 h. While oxidative stress was comparable, levels of circulating free DNA (cfDNA), and splenic/hepatic and pulmonary translocation of bacteria were decreased and increased, respectively at 48 h. DNases were increased compared to the established models. Proteomic analysis revealed an upregulation of systemic Epo, IL-1b, Prdx5, Parp1, Ccl2 and IL-6 at 48 h in comparison to the healthy controls. DISCUSSION AND CONCLUSION Midgut volvulus is a stable and physiological model for sepsis. Depending on the duration and subsequent tissue damage, it represents a combination of ischemia-reperfusion injury and hyperinflammation.
Collapse
Affiliation(s)
- Julia Elrod
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Antonia Kiwit
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Moritz Lenz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Daniela Börnigen
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christoph Mohr
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jasmin Knopf
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- Correspondence:
| |
Collapse
|
23
|
Intan PR, Noviantari A, Alegantina S. Response to Article "Combined Silver Sulfadiazine Nanosuspension with Thermosensitive Hydrogel: An Effective Antibacterial Treatment for Wound Healing in an Animal Model" [Letter]. Int J Nanomedicine 2023; 18:2257-2259. [PMID: 37152470 PMCID: PMC10162104 DOI: 10.2147/ijn.s408489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/27/2023] [Indexed: 05/09/2023] Open
Affiliation(s)
- Putri Reno Intan
- Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong - Bogor, West Java, Indonesia
- Correspondence: Putri Reno Intan, Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Genomic Building, Cibinong Science Center, Jalan Raya Bogor Km. 46, Cibinong – Bogor, West Java, 16911, Indonesia, Email
| | - Ariyani Noviantari
- Center for Biomedical Research, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong - Bogor, West Java, Indonesia
| | - Sukmayati Alegantina
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, Research Organization for Health, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong - Bogor, West Java, Indonesia
| |
Collapse
|
24
|
Linnemann C, Nussler AK, Histing T, Ehnert S. Febrile-Range Hyperthermia Can Prevent Toxic Effects of Neutrophil Extracellular Traps on Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:16208. [PMID: 36555846 PMCID: PMC9786713 DOI: 10.3390/ijms232416208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/30/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Fracture healing is characterized by an inflammatory phase directly after fracture which has a strong impact on the healing outcome. Neutrophils are strong contributors here and can release neutrophil extracellular traps (NETs). NETs are found after trauma, originally thought to capture pathogens. However, they can lead to tissue damage and impede wound healing processes. Their role in fracture healing remains unclear. In this study, the effect of isolated NETs on the function of bone-forming mesenchymal stem cells (SCP-1 cells) was examined. NETs were isolated from stimulated healthy neutrophils and viability, migration, and differentiation of SCP-1 cells were analyzed after the addition of NETs. NETs severely impaired the viability of SCP-1 cells, induced necrosis and already nontoxic concentrations reduced migration significantly. Short-term incubation with NETs had a persistent negative effect on osteogenic differentiation, as measured by AP activity and matrix formation. The addition of DNase or protease inhibitors failed to reverse the negative effect of NETs, whereas a short febrile-range temperature treatment successfully reduced the toxicity and membrane destruction. Thus, the possible modification of the negative effects of NETs in fracture hematomas could be an interesting new target to improve bone healing, particularly in patients with chronic diseases such as diabetes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
25
|
Zhong H, Lu RY, Wang Y. Neutrophil extracellular traps in fungal infections: A seesaw battle in hosts. Front Immunol 2022; 13:977493. [PMID: 36189199 PMCID: PMC9515532 DOI: 10.3389/fimmu.2022.977493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Fungal infections are a growing health care challenge. Neutrophils play a key role in defense against fungal infections. There are many effective ways for neutrophils to eliminate fungal invaders, such as phagocytosis, oxidative bursts, and the formation of extracellular traps. This process has received considerable attention and has made rapid progress since neutrophil extracellular traps (NETs) formation was described. Here, we describe the formation, induction, and function of NETs, as well as fungal strategies against NETs hunting. We highlight the effects of NETs on common fungal pathogens and how these pathogens survive.
Collapse
|
26
|
Koepke LG, Simon F, Heuer A. Neutrophil Extracellular Traps (NETs) im Gefäßsystem. GEFÄSSCHIRURGIE 2022; 27:444-448. [PMID: 36118259 PMCID: PMC9466318 DOI: 10.1007/s00772-022-00934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 12/01/2022]
Affiliation(s)
- L.-G. Koepke
- Klinik und Poliklinik für Unfallchirurgie und Orthopädie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Deutschland
| | - F. Simon
- Klinik für Gefäß- und Endovaskularchirurgie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Deutschland
| | - A. Heuer
- Klinik und Poliklinik für Unfallchirurgie und Orthopädie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Deutschland
- Mildred Scheel Nachwuchszentrum HaTriCS4, Universitäres Cancer Centrum Hamburg (UCCH), Hamburg, Deutschland
| |
Collapse
|
27
|
Mu R, Campos de Souza S, Liao Z, Dong L, Wang C. Reprograming the immune niche for skin tissue regeneration - From cellular mechanisms to biomaterials applications. Adv Drug Deliv Rev 2022; 185:114298. [PMID: 35439569 DOI: 10.1016/j.addr.2022.114298] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023]
Abstract
Despite the rapid development of therapeutic approaches for skin repair, chronic wounds such as diabetic foot ulcers remain an unaddressed problem that affects millions of people worldwide. Increasing evidence has revealed the crucial and diverse roles of the immune cells in the development and repair of the skin tissue, prompting new research to focus on further understanding and modulating the local immune niche for comprehensive, 'perfect' regeneration. In this review, we first introduce how different immunocytes and certain stromal cells involved in innate and adaptive immunity coordinate to maintain the immune niche and tissue homeostasis, with emphasis on their specific roles in normal and pathological wound healing. We then discuss novel engineering approaches - particularly biomaterials systems and cellular therapies - to target different players of the immune niche, with three major aims to i) overcome 'under-healing', ii) avoid 'over-healing', and iii) promote functional restoration, including appendage development. Finally, we highlight how these strategies strive to manage chronic wounds and achieve full structural and functional skin recovery by creating desirable 'soil' through modulating the immune microenvironment.
Collapse
|
28
|
Cutaneous Wound Healing: A Review about Innate Immune Response and Current Therapeutic Applications. Mediators Inflamm 2022; 2022:5344085. [PMID: 35509434 PMCID: PMC9061066 DOI: 10.1155/2022/5344085] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/22/2021] [Accepted: 03/25/2022] [Indexed: 12/22/2022] Open
Abstract
Skin wounds and compromised wound healing are major concerns for the public. Although skin wound healing has been studied for decades, the molecular and cellular mechanisms behind the process are still not completely clear. The systemic responses to trauma involve the body’s inflammatory and immunomodulatory cellular and humoral networks. Studies over the years provided essential insights into a complex and dynamic immunity during the cutaneous wound healing process. This review will focus on innate cell populations involved in the initial phase of this orchestrated process, including innate cells from both the skin and the immune system.
Collapse
|
29
|
Jiang Y, Xu X, Xiao L, Wang L, Qiang S. The Role of microRNA in the Inflammatory Response of Wound Healing. Front Immunol 2022; 13:852419. [PMID: 35386721 PMCID: PMC8977525 DOI: 10.3389/fimmu.2022.852419] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/20/2022] Open
Abstract
Wound healing, a highly complex pathophysiological response to injury, includes four overlapping phases of hemostasis, inflammation, proliferation, and remodeling. Initiation and resolution of the inflammatory response are the primary requirements for wound healing, and are also key events that determines wound quality and healing time. Currently, the number of patients with persistent chronic wounds has generally increased, which imposes health and economic burden on patients and society. Recent studies have found that microRNA(miRNA) plays an essential role in the inflammation involved in wound healing and may provide a new therapeutic direction for wound treatment. Therefore, this review focused on the role and significance of miRNA in the inflammation phase of wound healing.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Xiang Xu
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Long Xiao
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Lihong Wang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Sheng Qiang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| |
Collapse
|
30
|
Wang Z, Qi F, Luo H, Xu G, Wang D. Inflammatory Microenvironment of Skin Wounds. Front Immunol 2022; 13:789274. [PMID: 35300324 PMCID: PMC8920979 DOI: 10.3389/fimmu.2022.789274] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
Wound healing is a dynamic and highly regulated process that can be separated into three overlapping and interdependent phases: inflammation, proliferation, and remodelling. This review focuses on the inflammation stage, as it is the key stage of wound healing and plays a vital role in the local immune response and determines the progression of wound healing. Inflammatory cells, the main effector cells of the inflammatory response, have been widely studied, but little attention has been paid to the immunomodulatory effects of wound healing in non-inflammatory cells and the extracellular matrix. In this review, we attempt to deepen our understanding of the wound-healing microenvironment in the inflammatory stage by focusing on the interactions between cells and the extracellular matrix, as well as their role in regulating the immune response during the inflammatory stage. We hope our findings will provide new ideas for promoting tissue regeneration through immune regulation.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Plastic Surgery and Burns, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Fang Qi
- Department of Plastic Surgery and Burns, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Han Luo
- Department of Plastic Surgery and Burns, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Guangchao Xu
- Department of Plastic Surgery and Burns, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Dali Wang
- Department of Plastic Surgery and Burns, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
31
|
Zheng W, Zhu Z, Shi Y, Wen S, Ye L, Man M, Yang B, Liu Z. Neutrophils and their extracellular traps impair ablative fractional carbon dioxide laser-induced dermal remolding in mice. Lasers Surg Med 2022; 54:779-789. [PMID: 35181891 DOI: 10.1002/lsm.23526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/30/2022] [Accepted: 02/06/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Ablative fractional CO2 laser (AFL) therapy is an effective intervention to induce dermal remodeling. AFL treatment of the skin triggers the recruitment of immune cells, with neutrophils dominating the early phase. However, the role of recruited neutrophils in AFL-induced microinjuries and their subsequent dermal remodeling capacity remains elusive. MATERIALS AND METHODS A mouse model of AFL-induced dermal remodeling was established. RNA sequencing was used to identify the prominent features of AFL-treated tissues. Histological analysis, including H&E and Masson staining, ultrastructure observation by transmission microscopy, immunofluorescence, and quantitative real-time polymerase chain reaction were used for dermal remodeling analysis. Moreover, AFL-treated mice were intraperitoneally injected with anti-mouse Ly6G antibodies to deplete neutrophils. Neutrophil extracellular traps (NETs) were explored using immunofluorescence, transmission microscopy, and in vitro coculture experiments. RESULTS Dermal remodeling, characterized by an increased number of CD31-positve vessels and elevated messenger RNA (mRNA) expression of genes encoding transforming growth factor-β (TGF-β), collagen I, and collagen III, was observed at 15 days after AFL treatment. In the AFL-induced inflammation phase, RNA sequencing identified neutrophil chemotaxis, and degranulation genes were significantly enriched. Histology and immunofluorescence staining of human and mouse tissues harvested at Day 1 after AFL treatment revealed significant neutrophil infiltration surrounding thermal-induced microinjuries. Neutrophil depletion decreased the expression of stress-related genes such as S100A8 and S100A9 in the early phase following AFL treatment. Importantly, neutrophil depletion enhanced dermal remodeling at Day 15, as reflected by enrichment of the extracellular matrix and collagen biosynthesis genes based on RNA sequencing. Moreover, increased collagen I, collagen III, and TGF-β mRNA expression, increased cell proliferation, and vascularity were observed. Interestingly, NETs, which could be induced by AFL-treated fibroblasts in vitro, were identified in both human and mouse tissues on Day 1 after AFL treatment. CONCLUSIONS AFL-treated human and mouse skin recruited a large number of neutrophils. The neutrophil surge impaired dermal remodeling in mice. The microenvironment and fibroblast functional modulation mediated by neutrophil degranulation and NET formation were determined to be the underlying mechanisms. Our results indicate that modification of infiltrated neutrophil activity might be a potential therapeutic target for AFL-induced dermal remodeling.
Collapse
Affiliation(s)
- Wenyue Zheng
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Zhenlai Zhu
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yanqiang Shi
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Si Wen
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Li Ye
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Maoqiang Man
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Zhenfeng Liu
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
32
|
Laggner M, Lingitz MT, Copic D, Direder M, Klas K, Bormann D, Gugerell A, Moser B, Radtke C, Hacker S, Mildner M, Ankersmit HJ, Haider T. Severity of thermal burn injury is associated with systemic neutrophil activation. Sci Rep 2022; 12:1654. [PMID: 35102298 PMCID: PMC8803945 DOI: 10.1038/s41598-022-05768-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Burn injuries elicit a unique and dynamic stress response which can lead to burn injury progression. Though neutrophils represent crucial players in the burn-induced immunological events, the dynamic secretion pattern and systemic levels of neutrophil-derived factors have not been investigated in detail so far. Serum levels of neutrophil elastase (NE), myeloperoxidase (MPO), citrullinated histone H3 (CitH3), and complement factor C3a were quantified in burn victims over 4 weeks post injury. Furthermore, the potential association with mortality, degree of burn injury, and inhalation trauma was evaluated. In addition, leukocyte, platelet, neutrophil, and lymphocyte counts were assessed. Lastly, we analyzed the association of neutrophil-derived factors with clinical severity scoring systems. Serum levels of NE, MPO, CitH3, and C3a were remarkably elevated in burn victims compared to healthy controls. Leukocyte and neutrophil counts were significantly increased on admission day and day 1, while relative lymphocytes were decreased in the first 7 days post burn trauma. Though neutrophil-derived factors did not predict mortality, patients suffering from 3rd degree burn injuries displayed increased CitH3 and NE levels. Accordingly, CitH3 and NE were elevated in cases with higher abbreviated burn severity indices (ABSI). Taken together, our data suggest a role for neutrophil activation and NETosis in burn injuries and burn injury progression. Targeting exacerbated neutrophil activation might represent a new therapeutic option for severe cases of burn injury.
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Marie-Therese Lingitz
- Division of General Anesthesia and Intensive Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Martin Direder
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Katharina Klas
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefan Hacker
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Landesklinikum Wiener Neustadt, 2700, Wiener Neustadt, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria.
| | - Thomas Haider
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
33
|
Zahn KB, Franz AM, Schaible T, Rafat N, Büttner S, Boettcher M, Wessel LM. Small Bowel Obstruction After Neonatal Repair of Congenital Diaphragmatic Hernia-Incidence and Risk-Factors Identified in a Large Longitudinal Cohort-Study. Front Pediatr 2022; 10:846630. [PMID: 35656380 PMCID: PMC9152166 DOI: 10.3389/fped.2022.846630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE In patients with a congenital diaphragmatic hernia (CDH), postoperative small bowel obstruction (SBO) is a life-threatening event. Literature reports an incidence of SBO of 20% and an association with patch repair and ECMO treatment. Adhesions develop due to peritoneal damage and underly various biochemical and cellular processes. This longitudinal cohort study is aimed at identifying the incidence of SBO and the risk factors of surgical, pre-, and postoperative treatment. METHODS We evaluated all consecutive CDH survivors born between January 2009 and December 2017 participating in our prospective long-term follow-up program with a standardized protocol. RESULTS A total of 337 patients were included, with a median follow-up of 4 years. SBO with various underlying causes was observed in 38 patients (11.3%) and significantly more often after open surgery (OS). The majority of SBOs required surgical intervention (92%). Adhesive SBO (ASBO) was detected as the leading cause in 17 of 28 patients, in whom surgical reports were available. Duration of chest tube insertion [odds ratio (OR) 1.22; 95% CI 1.01-1.46, p = 0.04] was identified as an independent predictor for ASBO in multivariate analysis. Beyond the cut-off value of 16 days, the incidence of serous effusion and chylothorax was higher in patients with ASBO (ASBO/non-SBO: 2/10 vs. 3/139 serous effusion, p = 0.04; 2/10 vs. 13/139 chylothorax, p = 0.27). Type of diaphragmatic reconstruction, abdominal wall closure, or ECMO treatment showed no significant association with ASBO. A protective effect of one or more re-operations has been detected (RR 0.16; 95% CI 0.02-1.17; p = 0.049). CONCLUSION Thoracoscopic CDH repair significantly lowers the risk of SBO; however, not every patient is suitable for this approach. GoreTex®-patches do not seem to affect the development of ASBO, while median laparotomy might be more favorable than a subcostal incision. Neonates produce more proinflammatory cytokines and have a reduced anti-inflammatory capacity, which may contribute to the higher incidence of ASBO in patients with a longer duration of chest tube insertion, serous effusion, chylothorax, and to the protective effect of re-operations. In the future, novel therapeutic strategies based on a better understanding of the biochemical and cellular processes involved in the pathophysiology of adhesion formation might contribute to a reduction of peritoneal adhesions and their associated morbidity and mortality.
Collapse
Affiliation(s)
- Katrin B Zahn
- Department of Pediatric Surgery, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,ERNICA Centre, Mannheim, Germany
| | - Anna-Maria Franz
- Department of Pediatric Surgery, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,ERNICA Centre, Mannheim, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Neysan Rafat
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sylvia Büttner
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,ERNICA Centre, Mannheim, Germany
| | - Lucas M Wessel
- Department of Pediatric Surgery, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,ERNICA Centre, Mannheim, Germany
| |
Collapse
|
34
|
Neutrophil Extracellular Traps Exacerbate Ischemic Brain Damage. Mol Neurobiol 2021; 59:643-656. [PMID: 34748205 DOI: 10.1007/s12035-021-02635-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
Most acute strokes are ischemic, and subsequent neuroinflammation promotes further damage leading to cell death but also plays a beneficial role by promoting cellular repair. Neutrophils are forerunners to brain lesions after ischemic stroke and exert elaborate functions. While neutrophil extracellular traps (NETs) possess a fundamental antimicrobial function within the innate immune system under physiological circumstances, increasing evidence indicates that NETosis, the release process of NETs, occurs in the pathogenic process of stroke. In this review, we focus on the processes of NET formation and clearance, the temporal and spatial alterations of neutrophils and NETs after ischemic damage, and how NETs are involved in several stroke-related phenomena. Generally, NET formation and release processes depend on the generation of reactive oxygen species (ROS) and the activation of nuclear peptidylarginine deiminase-4 (PAD4). The acid-base environment, oxygen concentration, and iron ions around the infarct may also impact NET formation. DNase 1 has been identified as the primary degrader of NETs in serum, while reactive microglia are expected to inhibit the formation of NETs around ischemic lesions by phagocytosis of neutrophils. The neutrophils and NETs are present in the perivascular space ipsilateral to the infarct arising after ischemic damage, peaking between 1 and 3 days postischemia, but their location in the brain parenchyma remains controversial. After the ischemic injury, NETs are involved in the destruction of neurological function primarily by disrupting the blood-brain barrier and promoting thrombosis. The potential effects of NETs on various ischemic nerve cells need to be further investigated, especially in the chronic ischemic phase.
Collapse
|
35
|
Zhu S, Yu Y, Ren Y, Xu L, Wang H, Ling X, Jin L, Hu Y, Zhang H, Miao C, Guo K. The emerging roles of neutrophil extracellular traps in wound healing. Cell Death Dis 2021; 12:984. [PMID: 34686654 PMCID: PMC8536667 DOI: 10.1038/s41419-021-04294-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022]
Abstract
Delayed wound healing causes problems for many patients both physically and psychologically, contributing to pain, economic burden, loss of function, and even amputation. Although many factors affect the wound healing process, abnormally prolonged or augmented inflammation in the wound site is a common cause of poor wound healing. Excessive neutrophil extracellular trap (NET) formation during this phase may amplify inflammation and hinder wound healing. However, the roles of NETs in wound healing are still unclear. Herein, we briefly introduce NET formation and discuss the possible NET-related mechanisms in wound healing. We conclude with a discussion of current studies, focusing on the roles of NETs in diabetic and normoglycemic wounds and the effectiveness of NET-targeting treatments in wound healing.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Ren
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huilin Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Jin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|