1
|
Wang Y, Yin J, Yu C, Wu D, Chen Y, Han Q, Li S, Zhang R, Wang W, Xu J. Neutrophil extracellular traps activate STING signaling to promote dendritic cell-driven rejection after liver transplantation. Int Immunopharmacol 2025; 160:114763. [PMID: 40449271 DOI: 10.1016/j.intimp.2025.114763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 06/03/2025]
Abstract
PURPOSE Post-transplant immune rejection affects graft function. Interaction between neutrophil extracellular traps (NETs) with specific immune cells and the specific mechanism in liver transplantation were still unclear. METHOD Clinical patients RNA-Seq results were used for GSEA and KEGG analysis. C57BL/6 and C3H mouse models and clinical samples were use to describe the disease phenotype characteristics through multiple immunofluorescence, flow cytometry and etc. Cell co-culture experiments were performed to clarify the mechanism pathway process. RESULTS RNA-Seq results analysis indicated that the NETs formation pathway was upregulated. Animal models confirmed that in liver transplant immune rejection status the formation of NETs in situ and peripheral cells increased and the level of cell-free DNA (cf-DNA) in peripheral cells increased. Reactive oxygen species (ROS) as a predisposing factor for NETs accumulated more in immune rejection status and NETs are rich in mitochondrial DNA (mtDNA). NETs promote dendritic cell maturation through STING-related pathways. NETs formation increases in patients with liver transplant immune rejection and is positively correlated with disease severity. CONCLUSION We found that NETs can regulate dendritic cell maturation through STING-related pathways after liver transplantation, which may ultimately promote the occurrence of liver transplant rejection, providing a new perspective for clinical diagnosis, treatment and prevention of liver transplant rejection.
Collapse
Affiliation(s)
- Yan Wang
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jie Yin
- Basic Medicine School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chenjiang Yu
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Dongdong Wu
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yizhang Chen
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qi Han
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Shipeng Li
- Department of Hepatopancreaticobiliary Surgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Rui Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, National Health Commission Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
2
|
Zhang Z, Wang Y, Li T, Wang H. NETosis in myocardial ischemia-reperfusion injury: From mechanisms to therapies (Review). Biomed Rep 2025; 23:113. [PMID: 40420974 PMCID: PMC12105085 DOI: 10.3892/br.2025.1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
The present review describes the mechanisms of NETosis and its role in myocardial ischemia-reperfusion injury (MIRI), focusing on the release of neutrophil extracellular traps (NETs) by activated neutrophils. NETs, composed of depolymerized chromatin and granule proteins, are crucial for pathogen entrapment, infection control and immune regulation. However, NET formation, linked to neutrophil death (NETosis), exacerbates MIRI by promoting inflammation and tissue damage. To address therapeutic strategies for NETosis in MIRI, several potential clinically significant approaches were explored, including peptidylarginine deaminase 4 inhibition, DNase therapy, antioxidants, inflammation modulation, and antithrombotic treatments, which not only provide novel diagnostic biomarkers and therapeutic targets in MIRI, but are also expected to improve patient prognosis and advance the development of personalised medicine.
Collapse
Affiliation(s)
- Ziyang Zhang
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanxin Wang
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Tie Li
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Hongfeng Wang
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
3
|
Xia Y, Lan J, Yang J, Yuan S, Xie X, Du Q, Du H, Nie W, Jiang B, Zhao L, Cai Z, Zhang X, Xiong Y, Li Y, He R, Tao J. Saturated fatty acid-induced neutrophil extracellular traps contribute to exacerbation and biologic therapy resistance in obesity-related psoriasis. Cell Mol Immunol 2025; 22:597-611. [PMID: 40169704 DOI: 10.1038/s41423-025-01278-7] [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: 08/19/2024] [Accepted: 03/06/2025] [Indexed: 04/03/2025] Open
Abstract
Psoriasis patients who are obese tend to have serious clinical manifestations and poor responses to various biological agents in most cases. However, the mechanisms by which obesity exacerbates psoriasis remain enigmatic. In this study, we found that the abundance of systemic and localized cutaneous neutrophil extracellular traps (NETs) associated with the obesity-induced aggravation of psoriasis was positively correlated with disease severity and that the inhibition of NETs alleviated psoriatic dermatitis in obese mice. Mechanistically, we found that changes in fatty acid composition in obese subjects resulted in the deposition of saturated fatty acids (SFAs), which promoted the release of NETs via the TLR4-MD2/ROS signaling pathway. We further revealed that NETs potentiate IL-17 inflammation, especially γδT17-mediated immune responses, in obesity-exacerbated psoriasis patients. Moreover, SFAs induced a decreased response to anti-IL17A treatment in psoriasis-like mice, whereas the inhibition of NETs improved the beneficial effects of anti-IL17A in psoriasis-like mice with lipid metabolism disorders. Our findings collectively suggest that SFA-induced NETs play a critical role in the exacerbation of obesity-related psoriasis and provide potential new strategies for the clinical treatment of refractory psoriasis patients with lipid metabolism disorders.
Collapse
Affiliation(s)
- Yuting Xia
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajia Lan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Jing Yang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Shijie Yuan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaorong Xie
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyang Du
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyao Du
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Wenjia Nie
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Biling Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Liang Zhao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Zhen Cai
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Xin Zhang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Yan Xiong
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Yan Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China
| | - Ran He
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Gao F, Peng H, Gou R, Zhou Y, Ren S, Li F. Exploring neutrophil extracellular traps: mechanisms of immune regulation and future therapeutic potential. Exp Hematol Oncol 2025; 14:80. [PMID: 40442839 PMCID: PMC12123823 DOI: 10.1186/s40164-025-00670-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
Neutrophil extracellular traps (NETs) are complex, web-like structures consisting of DNA intertwined with antimicrobial proteins, which neutrophils release upon immune activation. These structures play a crucial role in pathogen elimination, particularly in infectious diseases. However, their involvement in various pathological conditions is multifaceted and context-dependent, while NETs contribute to host defense against infections, they can also exacerbate sterile inflammation, autoimmune disorders, and tumor progression. This review provides a comprehensive analysis of the molecular mechanisms governing NET formation and examines their interactions with immune cells, emphasizing how these interactions shape immune responses and drive disease dynamics. Furthermore, it explores ongoing clinical trials and emerging therapeutic strategies targeting NETs, offering critical insights into their potential translational applications in clinical practice.
Collapse
Affiliation(s)
- Fan Gao
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ruixue Gou
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yulan Zhou
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China
| | - Simei Ren
- National Center for Clinical Laboratories, Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Fei Li
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China.
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, China.
| |
Collapse
|
5
|
Gao F, Pan L, Liu W, Chen J, Wang Y, Li Y, Liu Y, Hua Y, Li R, Zhang T, Zhu T, Jin F, Gao Y. Idiopathic pulmonary fibrosis microenvironment: Novel mechanisms and research directions. Int Immunopharmacol 2025; 155:114653. [PMID: 40222273 DOI: 10.1016/j.intimp.2025.114653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive interstitial lung disease marked by increasing dyspnea and respiratory failure. The underlying mechanisms remain poorly understood, given the complexity of its pathogenesis. This review investigates the microenvironment of IPF to identify novel mechanisms and therapeutic avenues. Studies have revealed that various cell types, including alveolar epithelial cells, fibroblasts, myofibroblasts, and immune cells, are integral to disease progression, engaging in cellular stress responses and inflammatory regulation via signaling pathways such as TGF-β, Wnt, mTOR, and ROS. Non-coding RNAs, particularly miRNAs, are critical in IPF and may serve as diagnostic and prognostic biomarkers. Regarding treatment, mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) or non-vesicular derivatives offer promise by modulating immune responses, enhancing tissue repair, and inhibiting fibrosis. Additionally, alterations in the lung microbiota are increasingly recognized as a contributing factor to IPF progression, offering fresh insights into potential treatments. Despite the encouraging results of MSC-based therapies, the precise mechanisms and clinical applications remain subjects of ongoing research. This review emphasizes the significance of the IPF microenvironment and highlights the need for further exploration to develop effective therapies that could enhance patient outcomes.
Collapse
Affiliation(s)
- Fuguo Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wei Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jian Chen
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yifeng Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China; Department of Pulmonary and Critical Care Medicine, Shaanxi provincal people's hospital, Xi'an, 710068, China
| | - Yurou Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yiying Hua
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ruiqi Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Tongtong Zhang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ting Zhu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Yongheng Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
6
|
Yin Y, Chen Z, Yang C, Du H, Jiang G, Ji DK, Dai J. Spatiotemporally Controlled Co-Treatment of Lung Cancer and Pulmonary Immune-Related Adverse Events by Inhalable Albumin-Chaperoned Ultrathin MnO 2 Nanosheets. ACS NANO 2025. [PMID: 40378311 DOI: 10.1021/acsnano.5c01888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Cancer immunotherapy is often associated with immune-related adverse events (irAE), particularly in the lungs, which can lead to treatment discontinuation and negatively impact patient outcomes. In this study, we explore a spatiotemporally controlled cotreatment of lung cancer and pulmonary irAE using inhalable albumin-chaperoned ultrathin MnO2 nanosheets (MnO2@BSA). The MnO2@BSA exhibits efficient reactive oxygen species scavenging and NIR-II photothermal properties. The inhalation route enhances MnO2@BSA accumulation in the lungs and tumor tissue, offering localized treatment with reduced systemic toxicity. We demonstrate that ultrathin MnO2@BSA nanosheets can inhibit the formation of neutrophil extracellular traps (NETs) by reducing reactive oxygen species (ROS) in neutrophils, thereby alleviating inflammation associated with irAE. In animal models, inhaled MnO2@BSA reduced lung injury and inflammatory cell infiltration, while also decreasing pro-inflammatory cytokine levels, such as TNF-α, IL-1β, and IL-6. Simultaneously, MnO2@BSA displays strong photothermal properties under 1064 nm laser irradiation, effectively ablating tumors. This photothermal therapy also induces immunogenic cell death (ICD), promoting the reuse of activated CD8+ T cells to enhance antitumor immunity. These dual effects─tumor destruction and irAE mitigation─highlight MnO2@BSA's potential as a therapeutic platform for addressing the challenges of immunotherapy in lung cancer. We anticipate that this work could contribute to the development of inhalable metal-based nanomedicine for clinical transformation in tumor immunotherapy.
Collapse
Affiliation(s)
- Yanze Yin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhimin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Changjie Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Han Du
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ding-Kun Ji
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Jie Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
7
|
Tao P, Su B, Mao X, Lin Y, Zheng L, Zou X, Yang H, Liu J, Li H. Interleukin-35 inhibits NETs to ameliorate Th17/Treg immune imbalance during the exacerbation of cigarette smoke exposed-asthma via gp130/STAT3/ferroptosis axis. Redox Biol 2025; 82:103594. [PMID: 40101533 PMCID: PMC11964675 DOI: 10.1016/j.redox.2025.103594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
Cigarette smoke (CS) exposure amplifies neutrophil accumulation. IL-35, a novel cytokine with anti-inflammatory properties, is involved in protection against asthma. However, the biological roles of neutrophils and the precise molecular mechanisms of IL-35 in CS exposed-asthma remain unclear. We showed that the exacerbation of CS exposed-asthma leads to dramatically increased neutrophil counts and an imbalance in DC-Th17/Treg immune responses. RNA sequencing revealed that NETs, part of a key biological process in neutrophils, were significantly upregulated in the context of CS exposed-asthma exacerbation and that IL-35 treatment downregulated NET-associated gene expression. Targeted degradation of NETs, rather than neutrophil depletion, alleviated the CS exposed-asthma. Mechanistically, STAT3 phosphorylation promoted ferroptosis, exacerbating NET release, which in turn enhanced dendritic cell (DC) antigen presentation, activated T cells, and specifically promoted Th17 cell differentiation while inhibiting Treg cells. IL-35 acting on the gp130 receptor alleviated STAT3-mediated ferroptosis-associated NET formation. In summary, our study revealed a novel mechanism by which IL-35 inhibited NET formation, subsequently alleviating neutrophilic inflammation and restoring the DC-Th17/Treg imbalance in CS exposed-asthma, highlighting the potential of IL-35 as a targeted therapeutic strategy.
Collapse
Affiliation(s)
- Peizhi Tao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Beiting Su
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xueyan Mao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yusen Lin
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Zheng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hailing Yang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Liu
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Hongtao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
8
|
Zhang J, Miao C, Zhang H. Targeting neutrophil extracellular traps in cancer progression and metastasis. Theranostics 2025; 15:5846-5869. [PMID: 40365275 PMCID: PMC12068306 DOI: 10.7150/thno.111096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Neutrophils serve as pivotal effectors and regulators of the intricate immune system. Their contributions are indispensable, encompassing the obliteration of pathogens and a significant role in both cancer initiation and progression. Conversely, malignancies profoundly affect neutrophil activity, maturation, and lifespans. Cancer cells manipulate their biology to enhance or suppress the key functions of neutrophils. This manipulation is one of the most remarkable defensive mechanisms used by neutrophils, including the formation of neutrophil extracellular traps (NETs). NETs are filamentous structures comprising DNA, histones, and proteins derived from cytotoxic granules. In this review, we discuss the bidirectional interplay in which cancer elicits NET formation, and NETs concurrently facilitate cancer progression. Here, we discuss how vascular dysfunction and thrombosis induced by neutrophils and NETs contribute to an elevated risk of mortality from cardiovascular complications in patients with cancer. Ultimately, we propose a series of therapeutic strategies that hold promise for effectively targeting NETs in clinical settings.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| |
Collapse
|
9
|
Huang Z, Li Z, Ruan D, Xu Y, Cai H, Liu H, Jin H, He P, Fei Y, Huang J, Wang C, Chen X, Jiang J, Shen W. Dynamic changes of molecular pattern and cellular subpopulation in puncture-induced tendon injury model. iScience 2025; 28:112034. [PMID: 40230536 PMCID: PMC11994932 DOI: 10.1016/j.isci.2025.112034] [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: 12/20/2023] [Revised: 07/03/2024] [Accepted: 02/12/2025] [Indexed: 04/16/2025] Open
Abstract
Tendon degeneration and injury often result in significant pain and functional impairment. Typically, tendon healing occurs through a scar-mediated response and may progress to chronic tendinopathy without timely intervention. However, the molecular mechanisms underlying early tendon repair remain poorly understood. Further investigation is also impeded by the limited availability of early tendon injury samples in clinical settings. In this study, we established a puncture-induced tendon injury model to investigate the molecular patterns and cellular subpopulations involved in early tendon injury across multiple time points. RNA sequencing identified seven gene sets with distinct expression profiles during the early stages of tendon injury. Single-cell RNA sequencing further revealed eight myeloid cell types and seven mesenchymal cell types participating in the tendon repair process. Together, these findings illuminate the molecular and cellular dynamics coordinating early tendon repair, providing insights that could inform future clinical treatments for tendinopathy and tendon injury.
Collapse
Affiliation(s)
- Zizhan Huang
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Ziyang Li
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Dengfeng Ruan
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Yiwen Xu
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Honglu Cai
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Hengzhi Liu
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Haocheng Jin
- Department of Orthopedics, National Center for Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Peiwen He
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Yang Fei
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Jiayun Huang
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Canlong Wang
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
| | - Xiao Chen
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, P.R. China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou City, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou City, Zhejiang Province, P.R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Jia Jiang
- Department of Orthopedics, National Center for Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Weiliang Shen
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, P.R. China
- Institute of Sports Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, P.R. China
- Zhejiang Key Laboratory of Motor System Disease Precision Research and Therapy, Hangzhou City, Zhejiang Province, P.R. China
- Clinical Research Center of Motor System Disease of Hangzhou City, Hangzhou City, Zhejiang Province, P.R. China
- Dr. Li Dak Sum-Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, P.R. China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou City, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou City, Zhejiang Province, P.R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| |
Collapse
|
10
|
Wei Z, Jiang Y, Gao X, Qian Y, Zhang D, Li Q, Liu Q. Neospora caninum triggers the release of heterophil extracellular traps in chickens. Vet Parasitol 2025; 337:110471. [PMID: 40245810 DOI: 10.1016/j.vetpar.2025.110471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Neospora caninum (N. caninum) has been recognized as a pathogen responsible for neosporosis in chicken. Heterophils, which are a crucial part of the chicken's innate immune response, employ heterophil extracellular traps (HETs) as a principal defense strategy against invading pathogens. However, whether N. caninum can trigger the release of HETs remains unclear. This study presents the first evidence that N. caninum can induce the release of HETs in chickens and delves into the mechanisms that regulate this phenomenon. The impact of N. caninum on heterophil activity was evaluated using the CCK-8 assay. N. caninum- induced HETs were characterized using scanning electron microscopy and immunofluorescence. The release of N. caninum-induced HETs was quantified using a PicoGreen-based fluorescence microplate assay. Our findings revealed that heterophil activity remained unaffected by N. caninum infection. N. caninum induced thicknesses, reminiscent of HET-like structures, which primarily consisted of a DNA skeleton, citrullinated histone (citH3), and elastin. Furthermore, an increase in reactive oxygen species (ROS) production was detected during the release of N. caninum-induced HETs, a process that was found to be dependent on ROS-mediated activation, NADPH oxidase activity, glycolysis, toll-like receptor 2/4 (TLR2/4) and MAPK signaling pathways. N. caninum induced-HETs release was shown to be ROS-dependent, involving the activation of NADPH oxidase, ERK1/2, glycolysis, p38 MAPK, and toll-like receptor signaling pathways. In conclusion, this study is the first to elucidate the release mechanism of N. caninum-induced HETs in chicken, offering significant insights into the mechanisms underlying immune innate system against N. caninum infection.
Collapse
Affiliation(s)
- Zhengkai Wei
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China.
| | - Yuqian Jiang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528225, China
| | - Xinxin Gao
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528225, China
| | - Yuxiao Qian
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528225, China
| | - Dezhi Zhang
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Qianyong Li
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Quan Liu
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528225, China
| |
Collapse
|
11
|
Li Z, Lin J, Su Z, Zeng Y, Zhou Y, Li J, Yu W, Ye G, Zheng G, Xiao Z, Wu Y, Shen H, Xie Z. Neutrophil Extracellular Traps-Associated RNA Impedes CD4 + Treg Differentiation by TLR7-IRF7 Axis in Ankylosing Spondylitis. Arthritis Rheumatol 2025. [PMID: 40211099 DOI: 10.1002/art.43166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Our objective was to investigate the role of neutrophil extracellular traps (NETs) in the pathogenesis of inflammatory disorders in ankylosing spondylitis (AS). METHODS Local and circulating NETs levels were determined by immunofluorescence (IF) and myeloperoxidase (MPO)-DNA quantification in both patients with AS and AS model SKG mice. Flow cytometry (FCM) was performed to detect the effect of NETs on CD4+ subpopulation differentiation. The therapeutic effects of the neutrophil elastase inhibitor sivelestat (SVT) and the peptidylarginine deiminase 4 (PAD4) inhibitor CI-amidine were evaluated in SKG mice. The localization of NETs and their ability to impede CD4+ Treg cell differentiation were evaluated via IF, FCM, and Western blotting. RNA sequencing and specific inhibitors were used to clarify the detailed mechanism by which NETs inhibit CD4+ Treg differentiation. RESULTS The NETs levels were elevated locally and systemically in both patients with AS and SKG mice, which impeded the differentiation of CD4+ Treg cells. Blocking NETs formation via SVT or CI-amidine restored the CD4+ Treg ratio and subsequently alleviated inflammation in SKG mice. NETs were internalized by CD4+ T cells, and their associated RNA activated the Toll-like receptor 7 (TLR7)-interferon regulatory factor 7 (IRF-7) axis, which then inhibited Treg differentiation. Inhibiting CD4+ T cells endocytosis, removing the bound RNA component, or blocking the TLR7-IRF-7 axis abrogated the negative effect of NETs on CD4+ Treg differentiation. CONCLUSION Elevated NETs impeded CD4+ Treg differentiation by activating the TLR7-IRF-7 axis via their associated RNA in AS, and targeting NETs may be a novel treatment strategy for AS and related inflammatory disorders.
Collapse
Affiliation(s)
- Zhikun Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, P. R. China
| | - Jiajie Lin
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Zepeng Su
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Yipeng Zeng
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, P. R. China
| | - Yi Zhou
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, P. R. China
| | - Jinteng Li
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Wenhui Yu
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Guiwen Ye
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Guan Zheng
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Zipeng Xiao
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, P. R. China
| | - Yanfeng Wu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, P. R. China
| | - Huiyong Shen
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| | - Zhongyu Xie
- The Eighth Affiliated Hospital, Sun Yat-sen University and Guangdong Provincial Clinical Research Center for Orthopedic Diseases, Shenzhen, P. R. China
| |
Collapse
|
12
|
Hou Y, Wang W, Ye J, Sun L, Zhou S, Zheng Q, Shi Y, Chen Y, Yao J, Wang L, Yan X, Wan R, Chen S, Li Y. The crucial role of neutrophil extracellular traps and IL-17 signaling in indomethacin-induced gastric injury in mice. Sci Rep 2025; 15:12109. [PMID: 40204883 PMCID: PMC11982219 DOI: 10.1038/s41598-025-95880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
The homeostasis of gastric mucosa is extremely delicate. Neutrophils, the most abundant immune cells in human circulation, are regarded crutial in the regulation of gastric mucosal immune response. Non-steroidal anti-inflammatory drugs (NSAIDs) induced gastric injury is the second major reason for gastric ulcers. The relations between neutrophils and Indomethacin-induced gastric injury are not fully understood. A mouse model of gastric injury was established using Indomethacin, followed by proteomic analysis (raw data are available via ProteomeXchange with identifier PXD058482). GO functional annotations and KEGG pathway enrichment analysis were conducted on significant differential proteins. The formation of neutrophil extracellular traps (NETs) was observed using ELISA and immunofluorescence. TEM, Western blot and Real-time PCR were applied to observe programmed death of gastric epithelial cells (GECs), and ELISA was conducted to measure levels of TNF-α and IL-1β in the gastric tissue. Deoxyribonuclease 1 (DNase 1), a NETs inhibitor, was administered intraperitoneally to inhibit NETs formation. In vitro, neutrophils were isolated from peripheral blood of mice and co-cultured with mouse GECs cell line, different dosage of Indomethacin were added to the culture dish, the levels of inflammatory factors, formation of NETs and GECs programmed death were assessed in vitro. Poly morphonuclear neutrophils (PMN) were extracted from mouse peripheral blood and single-cell RNA-sequencing (scRNA-seq) was further applied (raw data are available via Genome Sequence Archive with identifier CRA020950) to explore the intracellular mechanism of NETs formation. ELISA and immunofluorescence were performed to validate expression of IL-17 signaling pathway. After Indomethacin gavage, obvious gastric injury was observed. Proteomic analysis indicated that NETs formation played a crucial role in Indomethacin-induced gastric injury. Compared to control group, Indomethacin treatment resulted in NETs formation, elevated levels of TNF-α and IL-1β and GECs programmed death. Inhibition of NETs significantly reduced inflammatory factor levels and mitigated gastric injury caused by indomethacin. In vitro, 200 µL, 400 µL and 600 µL of Indomethacin caused excessive NETs formation in neutrophils. Besides, Indomethacin-induced NETs formation led to GECs programmed death in vitro. scRNA-seq revealed that neutrophils enrichment in the peripheral blood of Indomethacin-induced gastric injury and IL-17 signaling might be the key intracellular of NETs formation. Expressions of neutrophil IL-17R and concentration of IL-17 were significantly higher in model group. NETs formation is pivotal in Indomethacin-induced gastric injury, contributing to programmed cell death of GECs and inflammation; IL-17 signaling might be the key intracellular mechanism of NETs formation.
Collapse
Affiliation(s)
- Yujun Hou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Wang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiangnan Ye
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luqiang Sun
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyuan Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianhua Zheng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunzhou Shi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junpeng Yao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Wang
- Department of Acupuncture and Moxibustion, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangyun Yan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renhong Wan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuai Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Li
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
13
|
Wu Y, Ning K, Huang Z, Chen B, Chen J, Wen Y, Bu J, Hong H, Chen Q, Zhang Z, Jia R, Su W. NETs-CD44-IL-17A Feedback Loop Drives Th17-Mediated Inflammation in Behçet's Uveitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411524. [PMID: 40013981 PMCID: PMC12021058 DOI: 10.1002/advs.202411524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Behçet's uveitis (BU) is a severe ocular manifestation of Behçet's disease, typically accompanied by abnormal neutrophil infiltration and hyperactivation. However, the underlying causes of excessive neutrophil extracellular traps (NETs) production and mechanisms by which NETs contribute to the pathogenesis of BU remain incompletely understood. Neutrophils from BU patients exhibit a higher propensity for NETs release compared to healthy controls. In the experimental autoimmune uveitis (EAU), neutrophils are observed to exert pro-inflammatory effects through NETs. Clearing NETs can inhibit T helper 17 (Th17) cell differentiation and significantly alleviate EAU symptoms. In vivo and in vitro experiments demonstrate neutralizing IL-17A markedly reducing neutrophil infiltration and NETs formation in EAU. Single-cell RNA sequencing confirms that CD44 plays a key role in mediating interactions between NETs and Th17 cells. Antagonizing CD44 inhibits the proportion of Th17 cells and NETs formation. Multiplex immunofluorescence and cell communication analyses further demonstrate interactions and colocalization between NETs and CD44highCD4+T cells in EAU. NETs induce Th17 differentiation via upregulating CD44, and in turn, Th17 cells secrete IL-17A to recruit neutrophils and promote NETs formation. Interrupting NETs-CD44-IL-17A feedback loop may be a potential therapeutic target for BU.
Collapse
Affiliation(s)
- Yi Wu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangdong Provincial Clinical Research Center for Ocular DiseasesGuangzhou510060China
| | - Kang Ning
- Department of Head and Neck SurgerySun Yat‐sen University Cancer CenterGuangzhou510050China
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510050China
| | - Zhaohao Huang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangdong Provincial Clinical Research Center for Ocular DiseasesGuangzhou510060China
| | - Binyao Chen
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangdong Provincial Clinical Research Center for Ocular DiseasesGuangzhou510060China
| | - Junjie Chen
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangdong Provincial Clinical Research Center for Ocular DiseasesGuangzhou510060China
| | | | - Jian Bu
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Han Hong
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Qiaorong Chen
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Zhuoqi Zhang
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Renbing Jia
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Wenru Su
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangdong Provincial Clinical Research Center for Ocular DiseasesGuangzhou510060China
- Department of OphthalmologyShanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| |
Collapse
|
14
|
Shen S, Fang H, Li X, Zhou Y, Tang X, Miao H, Li L, Chen J, Xue K, Zhang C, Chu M, Pang B, Bai Y, Qiao H, Dang E, Shao S, Wang G. Eosinophil extracellular traps drive T follicular helper cell differentiation via VIRMA-dependent MAF stabilization in bullous pemphigoid. J Allergy Clin Immunol 2025; 155:1357-1370. [PMID: 39490750 DOI: 10.1016/j.jaci.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Bullous pemphigoid (BP) is an autoimmune blistering disease characterized by the presence of pathogenic autoantibodies and a substantial influx of immune cells into skin lesions. However, the role of eosinophils in BP remains inadequately elucidated. OBJECTIVE We sought to determine the pathologic involvement of eosinophils and eosinophil extracellular traps (EETs) in BP. METHODS Human samples collected from BP patients and healthy controls were utilized to explore the potential role of eosinophils and their EETs in BP patients through serologic detection, flow cytometry, and immunofluorescence. Naive CD4+ T cells isolated from healthy donors were stimulated and subjected to further analysis via RNA sequencing. We additionally evaluated the potential of targeting EETs in BP180-immunized BP-like mice and in in vitro settings. RESULTS We found that elevated levels of eosinophils and EETs in BP patients correlated with disease severity. The DNA components within EETs played a crucial role in driving the differentiation of naive CD4+ T cells into follicular helper T (Tfh) cells by activating coil domains containing 25 (CCDC25). Treatment with DNase I, which disrupts the structural integrity of EETs, or neutralizing antibody against CCDC25 reduced the expansion of Tfh cells and suppressed the production of autoantibodies in BP180-immunized BP-like mouse models. Additionally, we discovered that EETs induced the N6-methyladenosine methylation of the transcription factor musculoaponeurotic fibrosarcoma (MAF) via the DNA-CCDC25-VIRMA pathway, thereby enhancing its mRNA stability and promoting Tfh cell differentiation. CONCLUSION Our study revealed a previously unrecognized mechanism by which EETs trigger abnormal Tfh cell differentiation through CCDC25, followed by Vir-like m6A methyltransferase-associated protein (VIRMA)-mediated N6-methyladenosine modification of MAF. These insights provide promising avenues for the development of targeted therapeutic interventions in the field of BP and potentially other autoimmune diseases.
Collapse
Affiliation(s)
- Shengxian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xia Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yifan Zhou
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Tang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haijun Miao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiaoling Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke Xue
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mengyang Chu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongjiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
15
|
Chen C, Zhang A, Cheng J, Yao Z, Meng J, Qin Y, Lu Q, Li Y, Liu X, Li T, Hou C, Tang Y, Liu H, Xu N, Dong S, Li X, Xu F, Guo J, Li C. Identification of Three Distinct Subgroups in Antiphospholipid Syndrome: Implication for Sex Differences and Prognostic Outcomes from a Multicenter Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415291. [PMID: 39965097 PMCID: PMC12005735 DOI: 10.1002/advs.202415291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Antiphospholipid syndrome (APS) is a heterogeneous autoimmune disease with persistent antiphospholipid antibodies. This study aimed to identify unrecognized APS subgroups from multicenter cohorts (n = 760, training: n = 415; validation: n = 345). Patients are stratified through unsupervised K-means clustering analysis. Prognostic outcomes are evaluated using Kaplan-Meier survival analyses. Proteomic analysis is conducted on primary APS patients (n = 36) and healthy controls (n = 12). Key molecule insulin-like growth factor 1 is validated using ELISA. Three clusters are identified. Cluster 1 (n = 320, 42.1%) is completely consisted of females (100%), with predominant occurrence of pregnancy morbidity (88.8%) but low incidences of thrombocytopenia (18.4%) and thrombosis (15.0%), and a favorable prognosis. Cluster 2 (n = 309, 40.7%) is predominantly female (99.4%) and characterized by high thrombosis (85.8%) and thrombocytopenia (46.6%), low pregnancy morbidity (13.6%), and poor prognosis. Cluster 3 (n = 131, 17.2%) is predominantly male (99.2%), exhibiting highest thrombosis (96.2%) and moderate thrombocytopenia (32.8%), with worst prognosis. Immunological and proteomic analyses clearly differentiated three clusters. This study reveals a distinct difference between obstetric and thrombotic APS, and a sex-based distinction within thrombotic APS. Three APS subgroups display unique clinical and molecular characteristics, and marked difference in prognostic outcomes.
Collapse
Affiliation(s)
- Chen Chen
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Ao Zhang
- School of Information and Communication EngineeringBeijing University of Posts and TelecommunicationsBeijing100876China
| | - Jianhui Cheng
- State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjingJiangsu210023China
| | - Zhongqiang Yao
- Department of Rheumatology and ImmunologyPeking University Third HospitalBeijing100191China
| | - Juan Meng
- Department of Rheumatology and ImmunologyBeijing Chaoyang Hospital Affiliated to Capital Medical UniversityBeijing100020China
| | - Yilu Qin
- Department of Rheumatology and ImmunologyAffiliated Xinxiang Central Hospital of Xinxiang Medical UniversityXinxiangHenan453000China
| | - Qingyi Lu
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Yufei Li
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Xiangjun Liu
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Tianhao Li
- Department of Biomedical InformaticsSchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Chao Hou
- Department of Biomedical InformaticsSchool of Basic Medical SciencesPeking University Health Science CenterBeijing100191China
| | - Yundi Tang
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Hongjiang Liu
- Department of Rheumatology and ImmunologyWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Ning Xu
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Sai Dong
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Xinxin Li
- State Key Laboratory of Neurology and Oncology Drug DevelopmentNanjingJiangsu210023China
| | - Fangmin Xu
- School of Information and Communication EngineeringBeijing University of Posts and TelecommunicationsBeijing100876China
| | - Jianping Guo
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| | - Chun Li
- Department of Rheumatology and ImmunologyPeking University People's HospitalBeijing100044China
| |
Collapse
|
16
|
Nigi L, Pedace E, Dotta F, Sebastiani G. Neutrophils in Type 1 Diabetes: Untangling the Intricate Web of Pathways and Hypothesis. Biomolecules 2025; 15:505. [PMID: 40305198 PMCID: PMC12025241 DOI: 10.3390/biom15040505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Neutrophils are increasingly recognized as key contributors to the pathogenesis of Type 1 Diabetes (T1D), yet their precise mechanistic role in disease onset and progression remains incompletely understood. While these innate immune cells reside in pancreatic tissue and support tissue homeostasis under physiological conditions, they can also drive tissue damage by triggering innate immune responses and modulating inflammation. Within the inflammatory milieu, neutrophils establish complex, bidirectional interactions with various immune cells, including macrophages, dendritic cells, natural killer cells, and lymphocytes. Once activated, they may enhance the innate immune response through direct or indirect crosstalk with immune cells, antigen presentation, and β-cell destruction or dysfunction. These mechanisms underscore the multifaceted and dynamic role of neutrophils in T1D, shaped by their intricate immunological interactions. Further research into the diverse functional capabilities of neutrophils is crucial for uncovering novel aspects of their involvement in T1D, potentially revealing new therapeutic targets to modulate disease progression.
Collapse
Affiliation(s)
- Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Erika Pedace
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
- Tuscany Centre for Precision Medicine, 53100 Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| |
Collapse
|
17
|
Wang Z, Jiao Y, Diao W, Shi T, Geng Q, Wen C, Xu J, Deng T, Li X, Zhao L, Gu J, Deng T, Xiao C. Neutrophils: a Central Point of Interaction Between Immune Cells and Nonimmune Cells in Rheumatoid Arthritis. Clin Rev Allergy Immunol 2025; 68:34. [PMID: 40148714 DOI: 10.1007/s12016-025-09044-3] [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] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease involving activation of the immune system and the infiltration of immune cells. As the first immune cells to reach the site of inflammation, neutrophils perform their biological functions by releasing many active substances and forming neutrophil extracellular traps (NETs). The overactivated neutrophils in patients with RA not only directly damage tissues but also, more importantly, interact with various other immune cells and broadly activate innate and adaptive immunity, leading to irreversible joint damage. However, owing to the pivotal role and complex influence of neutrophils in maintaining homoeostasis, the treatment of RA by targeting neutrophils is very difficult. Therefore, a comprehensive understanding of the interaction pathways between neutrophils and various other immune cells is crucial for the development of neutrophils as a new therapeutic target for RA. In this study, the important role of neutrophils in the pathogenesis of RA through their crosstalk with various other immune cells and nonimmune cells is highlighted. The potential of epigenetic modification of neutrophils for exploring the pathogenesis of RA and developing therapeutic approaches is also discussed. In addition, several models for studying cell‒cell interactions are summarized to support further studies of neutrophils in the context of RA.
Collapse
Affiliation(s)
- Zhaoran Wang
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yi Jiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenya Diao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tong Shi
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qishun Geng
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Chaoying Wen
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiahe Xu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Tiantian Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoya Li
- The Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100193, China
| | - Lu Zhao
- China-Japan Friendship Clinical Medical College, Capital Medical University, Beijing, 100029, China
| | - Jienan Gu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
- China-Japan Friendship Hospital Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tingting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Cheng Xiao
- China-Japan Friendship Clinical Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100029, China.
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
18
|
Hu X, Kang B, Wang M, Lin H, Liu Z, Zhang Z, Gu J, Mai Y, Guo X, Ma W, Yan H, Wang S, Huang J, Wang J, Zhang J, Zhang T, Feng B, Zhu Y, Pan G. Human induced pluripotent stem cells derived neutrophils display strong anti-microbial potencies. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:8. [PMID: 40113653 PMCID: PMC11926315 DOI: 10.1186/s13619-025-00227-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Neutrophils are essential innate immune cells with unusual anti-microbial properties while dysfunctions of neutrophils lead to severe health problems such as lethal infections. Generation of neutrophils from human induced pluripotent stem cells (hiPSCs) is highly promising to produce off-the-shelf neutrophils for transfusion therapies. However, the anti-microbial potencies of hiPSCs derived neutrophils (iNEUs) remain less documented. Here, we develop a scalable approach to generate iNEUs in a chemical defined condition. iNEUs display typical neutrophil characters in terms of phagocytosis, migration, formation of neutrophil extracellular traps (NETs), etc. Importantly, iNEUs display a strong killing potency against various bacteria such as K.pneumoniae, P.aeruginosa, E.coli and S.aureus. Moreover, transfusions of iNEUs in mice with neutrophil dysfunction largely enhance their survival in lethal infection of different bacteria. Together, our data show that hiPSCs derived neutrophils hold strong anti-microbial potencies to protect severe infections under neutrophil dysfunction conditions.
Collapse
Affiliation(s)
- Xing Hu
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Baoqiang Kang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Mingquan Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Huaisong Lin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Zhiyong Liu
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhishuai Zhang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Jiaming Gu
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yuchan Mai
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Xinrui Guo
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Wanli Ma
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Yan
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Shuoting Wang
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Jingxi Huang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Junwei Wang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jian Zhang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Tianyu Zhang
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Feng
- School of Biomedical Sciences, Faculty of Medicine, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine, The Chinese University of Hong Kong, Room 105A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, NT, Hong Kong SAR, China
| | - Yanling Zhu
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China.
| | - Guangjin Pan
- National Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Center for Development and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China.
| |
Collapse
|
19
|
Zhang S, Wen Q, Su S, Wang Y, Wang J, Xie N, Zhu W, Wen X, Di L, Lu Y, Xu M, Wang M, Chen H, Duo J, Huang Y, Wan D, Tao Z, Zhao S, Chai G, Hao J, Da Y. Peripheral immune profiling highlights a dynamic role of low-density granulocytes in myasthenia gravis. J Autoimmun 2025; 152:103395. [PMID: 40043622 DOI: 10.1016/j.jaut.2025.103395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Myasthenia gravis (MG) is an autoimmune neuromuscular disease marked by dysregulation of several immune cell populations. Here we explored peripheral immune landscape, particularly the role of low-density granulocytes (LDGs). METHODS Single-cell and bulk RNA sequencing analyzed peripheral immune cells from MG patients pre- (n = 4) and after treatment (n = 2), as well as healthy controls (n = 3). Flow cytometry was employed for validating LDG subsets, and various functional assays were conducted to assess their impact on T cell proliferation and differentiation, NET formation, and ROS production. RESULTS Single-cell analysis highlighted a shift towards inflammatory Th1/Th17/Tfh subsets, an intense interferon-mediated immune response, and an expansion of immature myeloid subsets in MG. Flow cytometry showed increased LDGs correlated with disease severity. Unlike myeloid-derived suppressor cells, MG LDGs do not restrict T cell proliferation but induce a pro-inflammatory Th1/Th17 response. They also display enhanced spontaneous neutrophil extracellular traps (NETs) formation and basal reactive oxygen species (ROS) production. LDGs decreased after intravenous immunoglobulin and increased after prolonged immunotherapy in minimal manifestation status (MM), with reduced pro-inflammatory activity. Bulk RNA sequencing revealed significant transcriptional differences in LDGs, especially in cell cycle and granule protein genes. CONCLUSION Peripheral immune profiling sheds light on the intricate role of LDGs in MG. These cells, as a distinct subtype of neutrophils with a proinflammatory phenotype, are notable increased in MG, exacerbating chronic inflammation. Furthermore, immunotherapy expanded LDGs but reduced their proinflammatory capacities. The complex interplay of LDGs in MG underscores their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shengyao Su
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yaye Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jingsi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nairong Xie
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenjia Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinmei Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Li Di
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yan Lu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hai Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianying Duo
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yue Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongshan Wan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Tao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shufang Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guoliang Chai
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Olsthoorn SEM, van Krimpen A, Hendriks RW, Stadhouders R. Chronic Inflammation in Asthma: Looking Beyond the Th2 Cell. Immunol Rev 2025; 330:e70010. [PMID: 40016948 PMCID: PMC11868696 DOI: 10.1111/imr.70010] [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: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Asthma is a common chronic inflammatory disease of the airways. A substantial number of patients present with severe and therapy-resistant asthma, for which the underlying biological mechanisms remain poorly understood. In most asthma patients, airway inflammation is characterized by chronic activation of type 2 immunity. CD4+ T helper 2 (Th2) cells are the canonical producers of the cytokines that fuel type 2 inflammation: interleukin (IL)-4, IL-5, IL-9, and IL-13. However, more recent findings have shown that other lymphocyte subsets, in particular group 2 innate lymphoid cells (ILC2s) and type 2 CD8+ cytotoxic T (Tc2) cells, can also produce large amounts of type 2 cytokines. Importantly, a substantial number of severe therapy-resistant asthma patients present with chronic type 2 inflammation, despite the high sensitivity of Th2 cells for suppression by corticosteroids-the mainstay drugs for asthma. Emerging evidence indicates that ILC2s and Tc2 cells are more abundant in severe asthma patients and can adopt corticosteroid-resistance states. Moreover, many severe asthma patients do not present with overt type 2 airway inflammation, implicating non-type 2 immunity as a driver of disease. In this review, we will discuss asthma pathophysiology and focus on the roles played by ILC2s, Tc2 cells, and non-type 2 lymphocytes, placing special emphasis on severe disease forms.
Collapse
Affiliation(s)
- Simone E. M. Olsthoorn
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Anneloes van Krimpen
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary MedicineErasmus MC University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
21
|
Wang M, Li Y, Li J, Yan B, Wang C, Zhang L, Lan F. New insights into the endotypes of chronic rhinosinusitis in the biologic era. J Allergy Clin Immunol 2025:S0091-6749(25)00211-8. [PMID: 39986619 DOI: 10.1016/j.jaci.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Chronic rhinosinusitis (CRS) exhibits significant heterogeneity and has been generally classified as type 1 (T1), T2, and T3 endotypes according to the histopathologic and inflammatory features of the nasal mucosa. T2 inflammation has been regarded as the predominant endotype of CRS linked to disease severity and refractory conditions. The development of biological agents that specifically target key molecules involved in T2 inflammation offers a highly effective and promising therapeutic approach for CRS. Recent findings have expanded the understanding of CRS endotypes by incorporating a range of disease-related molecules for classification, with progress made on the endotyping of CRS without nasal polyps. In addition, there has been an increasing emphasis on the study of mixed inflammatory endotypes. This review examines recent findings on CRS endotyping and the related noninvasive biomarkers, as well as novel mechanisms governing endotype formation, and addresses the efficacy of biologics in targeting T2 inflammation. Further research is warranted to understand if newly identified CRS endotypes show clinical significance for precision medicine and the management and treatment of refractory CRS in the era of biologics.
Collapse
Affiliation(s)
- Ming Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyun Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Bing Yan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China; Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.
| | - Feng Lan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of New Medicine and Diagnostic Technology Research for Nasal Disease, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China; Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
22
|
Zhang H, Guo R, Han Y, Yao Z, Quan M, Li B, Guo L. Alterations in neutrophil mRNA profiles in multiple sclerosis and identification of candidate genes for further investigation. Front Neurol 2025; 16:1548196. [PMID: 40035034 PMCID: PMC11873095 DOI: 10.3389/fneur.2025.1548196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Multiple sclerosis (MS) is a chronic and debilitating inflammatory disease of the central nervous system (CNS), characterized by demyelination and neurodegeneration. Emerging evidence implicates neutrophils in MS pathogenesis, particularly through processes like neutrophil extracellular traps (NETs) formation and degranulation, which may exacerbate inflammation and autoimmunity. Methods RNA sequencing of peripheral blood neutrophils from MS patients and healthy controls identified differentially expressed genes (DEGs). Pathway enrichment and protein-protein interaction (PPI) analyses highlighted potential biomarkers, validated using reverse transcription quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Results Our analysis identified 1,968 DEGs in neutrophils from MS patients, comprising 1,068 upregulated and 900 downregulated genes. Pathway enrichment analysis revealed significant involvement of immune processes, including antigen presentation, B and T cell receptor signaling, intracellular signaling cascades, and neutrophil degranulation. Notably, KEGG analysis highlighted a pivotal role for upregulated genes in neutrophil extracellular traps (NETs) formation, a process increasingly associated with autoimmunity. PPI network analysis pinpointed five key hub genes-LCN2, LTF, ELANE, CAMP, and CTSG-as central players in neutrophil-mediated immune modulation. Protein-level validation using ELISA confirmed elevated levels of LCN2, ELANE, CAMP, and CTSG, consistent with transcriptomic findings, further supporting their role as biomarkers. Subsequent RT-qPCR validation demonstrated robust diagnostic potential for these genes, with area under the curve (AUC) values of 0.952 (LCN2), 0.827 (LTF), 0.968 (ELANE), 0.950 (CAMP), and 0.862 (CTSG). Discussion These findings uncover a previously underappreciated role for neutrophils in MS pathogenesis, driven by alterations in gene expression linked to immune modulation and NET formation. The identified biomarkers, particularly ELANE and LCN2, demonstrate strong diagnostic potential, offering a new avenue for non-invasive MS diagnostics. Beyond clinical utility, this study highlights the importance of neutrophil-driven immune responses in MS, providing mechanistic insights into the complex interplay between innate and adaptive immunity in demyelinating diseases. Furthermore, these findings suggest that targeting neutrophil-specific processes, such as NETs formation and degranulation, could mitigate inflammatory damage and provide novel therapeutic approaches for MS treatment. These results lay the groundwork for future studies exploring therapeutic strategies targeting neutrophil functions in MS.
Collapse
Affiliation(s)
- Huining Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Ruoyi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yusen Han
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Zhichao Yao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Moyuan Quan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Clinical Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| |
Collapse
|
23
|
Mao JY, Xie YW, Lei XL, Zhang JH, Cheng W, Cui N. Effects of neutrophil granule proteins on sepsis-associated lymphopenia and their relationship with CD4 + T-cell pyroptosis. Front Immunol 2025; 16:1507800. [PMID: 39991146 PMCID: PMC11842378 DOI: 10.3389/fimmu.2025.1507800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Background Neutrophil acts as a double-edged sword in the immune system. We hypothesized that an elevated neutrophil granule protein level is associated with sepsis-associated lymphopenia (SAL). Methods We enrolled 61 patients with sepsis admitted to the Department of Critical Care Medicine of Peking Union Medical College Hospital between May 2022 and October 2023 in this study. Clinical and immunological parameters were recorded. Levels of neutrophil granule proteins, including myeloperoxidase (MPO) and neutrophil elastase (NE), and pyroptosis factors were examined. Results Levels of neutrophil granule proteins (MPO, 82.9 vs. 175.3, p < 0 <.0001; NE, 56.3 vs. 144.2, p < 0.0001) were significantly higher in patients with sepsis with lymphopenia. Neutrophil granule protein levels were independently associated with SAL risk (MPO: OR = 1.0841, 95% CI, 1.0020-1.1730; NE: OR = 1.0540, 95% CI, 1.0040-1.1065). The area under the curve of MPO levels predicting SAL occurrence was 0.939 (95% CI, 0.846-0.984), and that of NE was 0.950 (95% CI, 0.862-0.989). Furthermore, neutrophil granule proteins were significantly correlated with CD4+ T cell and its pyroptosis [MPO and CD4+ T cells (r = -0.4039, p < 0.0001), CD4+NLRP3 (r = 0.4868, p < 0.0001), NE and CD4+ T cells (r = -0.5140, p < 0.0001), and CD4+NLRP3 (r = 0.6513, p < 0.0001)]. Conclusion Increased levels of neutrophil granule proteins were significantly associated with SAL incidence, and a significant relationship between neutrophil granule proteins and the pyroptosis pathway of CD4+ T cells was revealed. Clinical trial registration chictr.org.cn identifier ChiCTR-ROC-17010750.
Collapse
Affiliation(s)
| | | | | | | | | | - Na Cui
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare
Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Chen Y, Mei E, Nan S, Chen X, Zhang P, Zhu Q, Lan D, Qi S, Wang Y. Fibrin aggravates periodontitis through inducing NETs formation from mitochondrial DNA. Oral Dis 2025; 31:577-588. [PMID: 39054859 DOI: 10.1111/odi.15073] [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: 04/01/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVES This study investigated the role of fibrin on neutrophil extracellular traps (NETs) formation from neutrophils and to elucidate the involvement of mitochondria in NETs formation during periodontitis. MATERIALS AND METHODS Plasminogen-deficient (Plg-/-) mice were employed to evaluate the effects of fibrin deposition on inflammation, bone resorption, and neutrophil infiltration in periodontal tissues. In addition, in vitro tests evaluated fibrin's impact on neutrophil-driven inflammation. Mitochondrial reactive oxygen species (mtROS) levels within neutrophils were quantified utilizing flow cytometry and immunofluorescence in vitro. Furthermore, the anti-inflammatory properties of the mtROS scavenger, Mito-TEMPO, were confirmed to regulate the NET formation in vitro and in vivo. RESULTS Plasminogen deficiency resulted in increased fibrin deposition, neutrophil infiltration, inflammatory factors concentration, and alveolar bone resorption in periodontal tissues. After neutrophils were treated by fibrin in vitro, the expression of inflammatory factors, the formation of mtROS, and NETs enriched in mitochondrial DNA (mtDNA) were upregulated, which were reversed by Mito-TEMPO in vitro. Moreover, Mito-TEMPO alleviated inflammation in Plg-/- mice. CONCLUSIONS This study showed that fibrin deposition in gingiva induced the NET formation in Plg-/- mice, in which the DNA in NETs was from mitochondria depending on increasing mtROS.
Collapse
Affiliation(s)
- Yinan Chen
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Enhua Mei
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Shunxue Nan
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Xueting Chen
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Pengye Zhang
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Qingyu Zhu
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Dongmei Lan
- Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Shengcai Qi
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Quan M, Zhang H, Deng X, Liu H, Xu Y, Song X. Neutrophils, NETs and multiple sclerosis: a mini review. Front Immunol 2025; 16:1487814. [PMID: 39935468 PMCID: PMC11810747 DOI: 10.3389/fimmu.2025.1487814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and degenerative autoimmune disease characterized by the activation of various inflammatory cells, leads to demyelination and neuronal injury. Neutrophils, often underestimated in MS, are gaining increased attention for their significant functions in MS patients and the experimental autoimmune encephalomyelitis (EAE) animal model. Neutrophils play multiple roles in mediating the pathogenesis of autoimmune diseases, and numerous studies suggest that neutrophils might have a crucial role through neutrophil extracellular trap (NET) formation. Studies on NETs in MS are still in their infancy. In this review, we discuss the clinical perspective on the linkage between neutrophils and MS or EAE, as well as the role of NETs in the pathogenesis of MS/EAE. Further, we analyze the potential mechanisms by which NETs contribute to MS, the protective effects of NETs in MS, and their value as targets for disease intervention. NET formation and/or clearance as a therapeutic approach for MS still requires research in greater depth.
Collapse
Affiliation(s)
- Moyuan Quan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Huining Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Xiaohong Deng
- Department of Rehabilitation Medicine, Beijing Zhongguancun Hospital, Beijing, China
| | - Huijia Liu
- Department of Internal Medicine, The Military Special Care Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Yanqiu Xu
- Department of Neurology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Xiujuan Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
26
|
LI X, ZHOU J, ZHAO C, ZHANG Y. [Research Progress of Tumor-associated Neutrophils
in the Occurrence and Development of Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:55-62. [PMID: 39988440 PMCID: PMC11848648 DOI: 10.3779/j.issn.1009-3419.2025.101.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Indexed: 02/25/2025]
Abstract
Lung cancer is the malignant tumor with the highest mortality rate worldwide. The tumor microenvironment (TME) is a key factor in the progression of lung cancer, composed of tumor cells, signaling molecules, fibroblasts, immune cells, etc. Among them, tumor associated neutrophil (TAN), as an important component of immune cells in the TME, plays multiple roles in tumor cell proliferation, invasion, angiogenesis, and metastasis due to its aberrant function, and is closely associated with poor prognosis. However, there are limited researches on the mechanism of TAN in lung cancer. This review aims to provide more scientific basis for studying the therapeutic targets of lung cancer and developing new drugs by elucidating the different subtypes of TAN and their mechanisms of action in the occurrence and development of lung cancer.
.
Collapse
|
27
|
Santacroce L, Topi S, Cafiero C, Palmirotta R, Jirillo E. The Role of the Immune Response to Helicobacter pylori Antigens and Its Relevance in Gastric Disorders. GASTROINTESTINAL DISORDERS 2025; 7:6. [DOI: 10.3390/gidisord7010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
Helicobacter pylori (H.p.) is a Gram-negative bacterium endowed with gastric tropism. H.p. infection is widely spread throughout the world, accounting for various pathologies, such as peptic ulcer, gastric cancer, mucosa-associated lymphoid tissue lymphoma, and extra-gastric manifestations. This bacterium possesses several virulence factors, e.g., lipopolysaccharides (LPS), the toxins CagA and VacA, and adhesins, which elicit a robust immune response during the initial phase of the infection. Of note, the lipid A moiety of the LPS exhibits a lower endotoxic potency than that of other LPSs, thus facilitating infection through a mechanism of immune escape. H.p. colonization of the gastric mucosa induces an initial protective immune response with innate immune cells, e.g., neutrophils, monocytes, and macrophages, which engulf and kill bacteria. Moreover, the same cells, along with gastric epithelial cells, secrete cytokines and chemokines, which recruit T cells [T helper (h)1 and Th17 cells] to the site of infection, thus leading to H.p. eradication. In a large subset of individuals, the perturbation of such an immune equilibrium leads to a harmful response, with an expansion of T regulatory (TREG) cells, which suppress the protective immune response. In fact, TREG cells, via the production of interleukin (IL)-10, downregulate Th1- and Th17-related cytokines, thus allowing H.p. survival and the perpetuation of inflammation. As far as the humoral immune response is concerned, B cells, upon H.p. stimulation, produce autoreactive antibodies, and IgG anti-Lex antibodies are harmful to the gastric mucosa. In this review, the structure and function of H.p. antigenic components and immune mechanisms elicited by this bacterium will be described in relation to gastric damage.
Collapse
Affiliation(s)
- Luigi Santacroce
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania
| | - Skender Topi
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania
| | | | - Raffaele Palmirotta
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
| |
Collapse
|
28
|
Amin A, Nikdoust F, Khorram S, Marashi SM, Ghanavati P, Ameri F, Akbarzadeh A, Hasanvand A, Khodakarim N. Dengue virus infection: how platelet-leukocyte crosstalk shapes thrombotic events and inflammation. Mol Biol Rep 2025; 52:119. [PMID: 39804486 DOI: 10.1007/s11033-025-10222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/02/2025] [Indexed: 05/02/2025]
Abstract
Dengue virus (DENV) poses a considerable threat to public health on a global scale, since about two-thirds of the world's population is currently at risk of contracting this arbovirus. Being transmitted by mosquitoes, this virus is associated with a range of illnesses and a small percentage of infected individuals might suffer from severe vascular leakage. This leakage leads to hypovolemic shock syndrome, generally known as dengue shock syndrome, organ failure, and bleeding complications. The severe form of this disease is believed to be, at least partially, associated with inflammatory and thrombotic states. These issues are significantly affected by the activation of platelets and leukocytes, as well as their interactions, which may influence its prognosis. The platelets present in a thrombus are able to attract leukocytes to the site of injury. The intricate process leads to the significant accumulation, activation, and migration of leukocytes, thereby promoting thrombotic events and triggering inflammatory responses. The occurrence of these events, combined with the direct viral infection of endothelial cells, leads to vascular endothelialitis, the disruption of cellular membranes, and the subsequent release of DAMPs. As a result, considerable damage occurs in the endothelium, which activates neutrophils and platelets; thisleads to their interaction and initiates the process of Netosis. Collectively, these processes exacerbate inflammatory and thrombotic conditions. In this respect, current research has focused on understanding whether effective anti-inflammatory protocols can prevent thrombotic events or, conversely, if efficient anticoagulant regimens may lead to a reduction in cytokine storms and tissue damage. This review article aims to illuminate the platelet leukocyte crosstalk, detailing the mechanisms through which platelets may play a role in the pathogenesis of DENV. The research outputs are particularly important in severe cases, in which case their interactions with leukocytes can exacerbate both inflammation and thrombosis in a mutually reinforcing manner.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Farahnaz Nikdoust
- Department of Cardiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, IR, Iran
| | | | | | - Pedram Ghanavati
- Department of Neurosurgery, Firouzgar Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Farshid Ameri
- Department of Infection, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ashkan Akbarzadeh
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Hasanvand
- Department of General Surgery, Lorestan University of Medical Science, Khorramabad, Iran
- Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Nastaran Khodakarim
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
- Department of medical oncology and hematology, Hazrat Rasoul Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Lee W, Ko SY, Akasaka H, Weigert M, Lengyel E, Naora H. Neutrophil extracellular traps promote pre-metastatic niche formation in the omentum by expanding innate-like B cells that express IL-10. Cancer Cell 2025; 43:69-85.e11. [PMID: 39753138 PMCID: PMC11732717 DOI: 10.1016/j.ccell.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 08/31/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Disseminated cancer cells in the peritoneal fluid often colonize omental fat-associated lymphoid clusters but the mechanisms are unclear. Here, we identify that innate-like B cells accumulate in the omentum of mice and women with early-stage ovarian cancer concomitantly with the extrusion of chromatin fibers by neutrophils called neutrophil extracellular traps (NETs). Studies using genetically modified NET-deficient mice, pharmacologic inhibition of NETs, and adoptive B cell transfer show that NETs induce expression of the chemoattractant CXCL13 in the pre-metastatic omentum, stimulating recruitment of peritoneal innate-like B cells that in turn promote expansion of regulatory T cells and omental metastasis through producing interleukin (IL)-10. Ex vivo studies show that NETs elicit IL-10 production in innate-like B cells by inactivating SHP-1, a phosphatase that inhibits B cell activation pathways, and by generating reactive oxygen species. These findings reveal that NETs alter immune cell dynamics in the pre-metastatic omentum, rendering this niche conducive for colonization.
Collapse
Affiliation(s)
- WonJae Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Song Yi Ko
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hironari Akasaka
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Melanie Weigert
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| | - Honami Naora
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Dragoni G, Ke BJ, Picariello L, Abdurahiman S, Ceni E, Biscu F, Mello T, Polvani S, Innocenti T, Spalart V, Milani S, D'Hoore A, Bislenghi G, Scaringi S, Verstockt B, De Hertogh G, Martinod K, Galli A, Matteoli G, Vermeire S. The Impact of Peptidyl Arginine Deiminase 4-Dependent Neutrophil Extracellular Trap Formation on the Early Development of Intestinal Fibrosis in Crohn's Disease. J Crohns Colitis 2025; 19:jjae121. [PMID: 39126198 DOI: 10.1093/ecco-jcc/jjae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/29/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND AIMS During early phases of inflammation, activated neutrophils extrude neutrophil extracellular traps (NETs) in a peptidyl arginine deiminase 4 (PAD4)-dependent manner, aggravating tissue injury and remodeling. In this study, we investigated the potential pro-fibrotic properties and signaling of NETs in Crohn's disease (CD). METHODS NETs and activated fibroblasts were labeled on resected ileum from CD patients by multiplex immunofluorescence staining. NETs-treated human primary intestinal fibroblasts were analyzed by bulk RNA sequencing to uncover cell signaling pathways, and by high-throughput imaging to assess collagen production and migratory activity. Consequentially, TLR2/NF-κB pathway was evaluated by transfection of CCD-18Co fibroblasts with an NF-κB-luciferase reporter plasmid, incorporating C29 to block TLR2 signaling. A chronic dextran sulfate sodium (DSS) mouse model was used to define the specific role of PAD4 deletion in neutrophils (MRP8-Cre, Pad4fl/fl). RESULTS Immunofluorescence showed spatial colocalization of NETs and activated fibroblasts in ileal ulcerations of CD patients. Transcriptomic analysis revealed upregulation of pro-fibrotic genes and activation of Toll-like receptor signaling pathways in NETs-treated fibroblasts. NETs treatment induced fibroblast proliferation, diminished migratory capability, and increased collagen release. Transfection experiments indicated a substantial increase in an NF-κB expression with NETs, whereas C29 led to decreased expression and release of collagen. In line, a significant reduction in collagen content was observed in the colon of MRP8-Cre, Pad4fl/fl mice subjected to chronic DSS colitis. CONCLUSIONS NETs potentially serve as an initial stimulus for pathological activation of fibroblasts within the intestine via the TLR2/NF-κB pathway. Given their early involvement in inflammation, inhibition of PAD4 might offer a strategy to modulate both inflammation and fibrogenesis in CD.
Collapse
Affiliation(s)
- Gabriele Dragoni
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
- Department of Gastroenterology, IBD Referral Center, Careggi University Hospital, Florence, Italy
| | - Bo-Jun Ke
- Laboratory for Mucosal Immunology, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Lucia Picariello
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Saeed Abdurahiman
- Laboratory for Mucosal Immunology, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Elisabetta Ceni
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Francesca Biscu
- Laboratory for Mucosal Immunology, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Tommaso Mello
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Simone Polvani
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Tommaso Innocenti
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
- Department of Gastroenterology, IBD Referral Center, Careggi University Hospital, Florence, Italy
| | - Valérie Spalart
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Stefano Milani
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
- Department of Gastroenterology, IBD Referral Center, Careggi University Hospital, Florence, Italy
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Gabriele Bislenghi
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Stefano Scaringi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Gert De Hertogh
- Laboratory of Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Andrea Galli
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
- Department of Gastroenterology, IBD Referral Center, Careggi University Hospital, Florence, Italy
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Feješ A, Šebeková K, Borbélyová V. Pathophysiological Role of Neutrophil Extracellular Traps in Diet-Induced Obesity and Metabolic Syndrome in Animal Models. Nutrients 2025; 17:241. [PMID: 39861371 PMCID: PMC11768048 DOI: 10.3390/nu17020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The global pandemic of obesity poses a serious health, social, and economic burden. Patients living with obesity are at an increased risk of developing noncommunicable diseases or to die prematurely. Obesity is a state of chronic low-grade inflammation. Neutrophils are first to be recruited to sites of inflammation, where they contribute to host defense via phagocytosis, degranulation, and extrusion of neutrophil extracellular traps (NETs). NETs are web-like DNA structures of nuclear or mitochondrial DNA associated with cytosolic antimicrobial proteins. The primary function of NETosis is preventing the dissemination of pathogens. However, neutrophils may occasionally misidentify host molecules as danger-associated molecular patterns, triggering NET formation. This can lead to further recruitment of neutrophils, resulting in propagation and a vicious cycle of persistent systemic inflammation. This scenario may occur when neutrophils infiltrate expanded obese adipose tissue. Thus, NETosis is implicated in the pathophysiology of autoimmune and metabolic disorders, including obesity. This review explores the role of NETosis in obesity and two obesity-associated conditions-hypertension and liver steatosis. With the rising prevalence of obesity driving research into its pathophysiology, particularly through diet-induced obesity models in rodents, we discuss insights gained from both human and animal studies. Additionally, we highlight the potential offered by rodent models and the opportunities presented by genetically modified mouse strains for advancing our understanding of obesity-related inflammation.
Collapse
Affiliation(s)
| | - Katarína Šebeková
- Institute of Molecular Biomedicine, Medical Faculty, Comenius University, 83303 Bratislava, Slovakia; (A.F.); (V.B.)
| | | |
Collapse
|
32
|
Guo H, Liang Q, Xue Z, Yang J, Chen P, Ji J, Li J, Guo G, Cao H, Sha X, Zhao R, Dong C, Gu Z. Neutrophil Extracellular Traps Participate in the Pathogenesis of Lupus Through S100A10-Mediated Regulatory T-Cell Differentiation and Functional Abnormalities. Eur J Immunol 2025; 55:e202451298. [PMID: 39508544 DOI: 10.1002/eji.202451298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024]
Abstract
In systemic lupus erythematosus (SLE), neutrophil dysregulation and neutrophil extracellular traps (NETs) formation contribute to disease pathogenesis, potentially worsening the autoimmune response. Although research indicates NETs' involvement in various autoimmune conditions, their relationship with regulatory T cells (Tregs) in SLE remains elusive. In this study, in vivo experiments were involved in administering NET injections to C57BL/6 and MRL/Ipr mice. In vitro, a co-culture system facilitated interaction between Tregs and NETs. Proteomic analysis elucidated NET composition, while RNA sequencing delineated their impact on Treg differentiation. We demonstrated that increased NET levels correlate inversely with Treg abundance in SLE patients, influencing both their proportion and functionality. NET administration reduced Treg levels and induced lupus-like symptoms in C57BL/6 mice, exacerbating symptoms in MRL/Ipr mice. DNase I treatment mitigated NET effects, restoring Treg levels and alleviating symptoms. RNA sequencing revealed altered gene expression in naïve CD4+ T cells exposed to NETs. Additionally, proteomic analysis showed S100A10 protein changes between SLE patients and healthy controls, hindering Treg differentiation. NETs influence TLR-4 of naïve CD4+ T cells via S100A10, thereby modulating Treg proportion and functionality. These findings highlight the critical role of NETs in Treg differentiation in SLE, suggesting that targeting NETs may provide a novel therapeutic approach.
Collapse
Affiliation(s)
- Hua Guo
- Graduate School, Dalian Medical University, Dalian, China
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Qian Liang
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Zhonghui Xue
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Junling Yang
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Pengyu Chen
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Juan Ji
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Jing Li
- Graduate School, Dalian Medical University, Dalian, China
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Genkai Guo
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Haixia Cao
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Xiaoqi Sha
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Rui Zhao
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
33
|
Wu Y, Park J, Le QV, Byun J, Choi J, Xu E, Lee J, Oh YK. NET formation-mediated in situ protein delivery to the inflamed central nervous system. Nat Commun 2024; 15:10747. [PMID: 39737919 PMCID: PMC11686318 DOI: 10.1038/s41467-024-54817-7] [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: 04/08/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Delivering protein drugs to the central nervous system (CNS) is challenging due to the blood-brain and blood-spinal cord barrier. Here we show that neutrophils, which naturally migrate through these barriers to inflamed CNS sites and release neutrophil extracellular traps (NETs), can be leveraged for therapeutic delivery. Tannic acid nanoparticles tethered with anti-Ly6G antibody and interferon-β (aLy6G-IFNβ@TLP) are constructed for targeted neutrophil delivery. These nanoparticles protect interferon-β from reactive oxygen species and preferentially accumulate in neutrophils over other immune cells. Upon encountering inflammation, neutrophils release the nanoparticles during NET formation. In the female mouse model of experimental autoimmune encephalomyelitis, intravenous administration of aLy6G-IFNβ@TLP reduce disease progression and restore motor function. Although this study focuses on IFNβ and autoimmune encephalomyelitis, the concept of hitchhiking neutrophils for CNS delivery and employing NET formation for inflamed site-specific nanoparticle release can be further applied for delivery of other protein drugs in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Junho Byun
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
35
|
Prendecki M, Gurung A, Pisacano N, Pusey CD. The role of neutrophils in ANCA-associated vasculitis. Immunol Lett 2024; 270:106933. [PMID: 39362307 DOI: 10.1016/j.imlet.2024.106933] [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: 07/24/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) is a group of rare systemic autoimmune diseases characterised by necrotising inflammation of small blood vessels and usually associated with circulating ANCA. The pathophysiology of AAV is complex, involving many aspects of the innate and adaptive immune system. Neutrophils are central to the pathogenesis of AAV as they are both the target of the autoantibody and effector cells mediating vascular injury. We describe mechanisms for ANCA induced activation of neutrophils, the pathogenic mechanisms by which this leads to endothelial cell injury, and how neutrophil crosstalk modulates other aspects of the immune system in AAV.
Collapse
Affiliation(s)
- Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom.
| | - Angila Gurung
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Noelle Pisacano
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
36
|
Lin L, Sun C, Ye Y, Zhu P, Pan K, Chen L. Transcriptome analysis revealed that ischemic post-conditioning suppressed the expression of inflammatory genes in lung ischemia-reperfusion injury. Front Genet 2024; 15:1425420. [PMID: 39655220 PMCID: PMC11625726 DOI: 10.3389/fgene.2024.1425420] [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: 05/17/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Ischemic post-conditioning (I-post C) is a recognized therapeutic strategy for lung ischemia/reperfusion injury (LIRI). However, the specific mechanisms underlying the lung protection conferred by I-post C remain unclear. This study aimed to investigate the protective mechanisms and potential molecular regulatory networks of I-post C on lung tissue. Methods Transcriptome analysis was performed on rat lung tissues obtained from Sham, ischemia-reperfusion (IR), and I-post C groups using RNA-seq to identify differentially expressed genes (DEGs). Subsequently, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and gene set enrichment analysis (GSEA) were conducted to elucidate significantly enriched pathways in the IR and I-post C groups. Additionally, protein-protein interaction (PPI) network analysis was carried out to examine associations among the DEGs. Pathological changes in lung tissues were assessed using hematoxylin-eosin (H&E) staining. The expression levels of CXCL1 and CXCL6 in the IR and I-post C groups were evaluated through immunofluorescence and Western blotting. Results Our results showed that I-post C significantly attenuated both pulmonary edema and inflammatory cell infiltration. Transcriptome analysis identified 38 DEGs in the I-post C group compared to the IR group, comprising 21 upregulated and 17 downregulated genes. Among these, seven inflammation-related DEGs exhibited co-expression patterns with the Sham and IR groups, with notable downregulation of Cxcl1 and Cxcl6. GO analysis primarily linked these DEGs to neutrophil activation, chemotaxis, cytokine activity, and CCR chemokine receptor binding. KEGG analysis revealed enriched pathways, including the IL-17, TNF, and NF-κB signaling pathways. GSEA indicated downregulation of neutrophil chemotaxis and the IL-17 signaling pathway, correlating with reduced expression of Cxcl1 and Cxcl6. Validation of Cxcl1 and Cxcl6 mRNA expression via immunofluorescence and Western blotting supported the RNA-seq findings. Furthermore, a PPI network was constructed to elucidate interactions among the 29 DEGs. Conclusions Through RNA-Seq analysis, we concluded that I-post C may reduce inflammation and suppress the IL-17 signaling pathway, thereby protecting against lung damage caused by LIRI, potentially involving neutrophil extracellular traps.
Collapse
Affiliation(s)
- Liangen Lin
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Congcong Sun
- Department of Scientific Research Center, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Yuanwen Ye
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Peng Zhu
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Keyue Pan
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| | - Linglong Chen
- Department of Emergency, Wenzhou People’s Hospital, The Third Affiliated to Shanghai University, Wenzhou, Zhejiang, China
| |
Collapse
|
37
|
Li W, Yang J. Single-cell and bulk RNA sequencing-based screening and identification of extracellular trap network-related genes in neutrophils in acute myocardial infarction. Medicine (Baltimore) 2024; 103:e40590. [PMID: 39809140 PMCID: PMC11596368 DOI: 10.1097/md.0000000000040590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/31/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The neutrophil-mediated generation of neutrophil extracellular traps (NETs) results in an augmented inflammatory response and cellular tissue injury during acute myocardial infarction (AMI). Through the analysis of public database information, we discovered and confirmed putative critical genes involved in NETs-mediated AMI. METHODS The AMI dataset GSE66360 and the single-cell dataset GSE163465 were downloaded from the Gene Expression Omnibus database. Key genes were screened by bioinformatics. Quantitative real-time PCR (qRT-PCR) was used to verify the key genes, and then a Mendelian randomization (MR) study was conducted on the basis of the genome-wide association study to determine the causal relationship between key genes and AMI. Dimensionality reduction clustering, pseudo-time series, and cell communication were performed on the single-cell dataset to analyze the key genes screened by bulk RNA sequencing and the dynamic evolution of NETs in the AMI process. Immunohistochemistry and Western blot were used to verify the key genes. RESULTS Six key genes, IL1β, S100A12, TLR2, CXCL1, CXCL2, and CCL4, were screened out through bioinformatics. qRT-PCR results showed that compared with the control group, the expression of 5 key genes was upregulated in the AMI group. In the MR study, CXCL1 and CCL4 were observed to have a causal relationship with AMI. Single-cell analysis showed that NETs-related genes CCL4, CXCL2, and IL1β were highly expressed. Combining single cells, qRT-PCR and MR, gene CCL4 was selected as the focus of the study. H9c2 cardiomyocytes simulated myocardial infarction under hypoxia, and the results showed that the expression of gene CCL4 was increased. The immunohistochemical results of gene CCL4 showed that the expression was upregulated in the AMI group. CONCLUSIONS We found 6 key genes related to NETs-mediated cell damage during AMI. The results of MR showed that CXCL1 and CCL4 were causally related to AMI. Combining single cells, qRT-PCR and MR, gene CCL4 may play an important role in the AMI process. Our results may provide some insights into neutrophil-mediated cell damage during AMI.
Collapse
Affiliation(s)
- Wei Li
- The Second Clinical Medical College of Bin Zhou Medical College, Shandong, China
| | - Jun Yang
- Yantai Yuhuangding Hospital, Shandong, China
| |
Collapse
|
38
|
Pu D, Wang P, Wang X, Tian Y, Gong H, Ma X, Li M, Zhang D. Focusing on non-responders to infliximab with ulcerative colitis, what can we do first and next? Int Immunopharmacol 2024; 141:112943. [PMID: 39191122 DOI: 10.1016/j.intimp.2024.112943] [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: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic immune-mediated inflammation of the colorectum, for which infliximab (IFX) is currently the mainstay of treatment. However, one-third of patients with UC still fail to benefit from the IFX therapy, and early exposure to IFX impairs the efficacy of other subsequent biologics. Therefore, personalized therapeutic system is urgently needed to assist in clinical decision-making and precision treatment. METHODS Four microarray datasets of colonic biopsies from UC patients treated with IFX were obtained from the GEO database to form the Training Cohort and Validation Cohort. Differentially expressed genes (DEGs) in Training Cohort were identified and enriched for GO, KEGG and immune cell infiltration analysis. A prediction model for IFX efficacy was developed based on the LASSO and Logistic regression. The predictive accuracy of the model was verified by the Validation Cohort, and the model-genes/proteins were validated by immunohistochemistry. Gene-drug, gene-ncRNA interaction analysis were performed to identify drugs or non-coding RNAs (ncRNAs) that potentially interacted with the model-genes. Homology Modeling and Molecular Docking were conducted to filter the optimal candidate as the subsequent adjuvant or alternative for IFX in predicted non-responders. At last, the down-regulation of the key model-gene/protein CYP24A1 by the drug candidate Deferasirox was verified by Western Blot and qRT-PCR Assay based on cellular experiments. RESULTS A total of 113 DEGs were identified in the Training Cohort, mainly enriched in inflammatory cell chemotaxis, migration, and response to molecules derived from intestinal microbiota. Activated pro-inflammatory innate immune cells, including neutrophils, M1 macrophages, activated dendritic cells and mast cells, were significantly enriched in colons of non-responders. The prediction model based on three model-genes (IFI44L, CYP24A1, and RGS1) exhibited strong predictive efficacy, with AUC values of 0.901 and 0.80 in the Training and Validation Cohorts, respectively. Higher expression of the three model-genes/proteins in colons of non-responders to IFX was confirmed by clinical colonic mucosal biopsies. 4 Drugs (Calcitriol, Lunacalcipol, Deferasirox, Telaprevir), 15 miRNAs and 66 corresponding lnRNAs interacting with model-genes were identified. The protein 3D structure of the key model-gene/protein (human-derived CYP24A1) was developed. Through the Molecular Docking and cellular experimental validation, Deferasirox, which significantly down-regulated both the RNA and protein expression of CYP24A1, was identified as the optimal adjuvant or alternative for IFX in predicted non-responders with UC. CONCLUSION This study developed a novel prediction model for pre-assessing the efficacy of IFX in patients with UC, as the first step towards personalized therapy. Meanwhile, drugs and non-coding RNAs were provided as potential candidates to develop the next-step precise treatment for the predicted non-responders. In particular, Defeasirox appears to hold promise as an adjuvant or alternative to IFX for the optimization of UC therapy.
Collapse
Affiliation(s)
- Dan Pu
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Pengfei Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xiang Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Yonggang Tian
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xueni Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Muyang Li
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China.
| |
Collapse
|
39
|
van Straalen KR, Piguet V, Gudjonsson JE. Hidradenitis suppurativa: key insights into treatment success and failure. J Clin Invest 2024; 134:e186744. [PMID: 39484718 PMCID: PMC11527436 DOI: 10.1172/jci186744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Affiliation(s)
- Kelsey R. van Straalen
- Laboratory for Experimental Immunodermatology, Department of Dermatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Vincent Piguet
- Division of Dermatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Division of Dermatology, Department of Medicine, Women’s College Hospital, Toronto, Canada
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Taubman Medical Research Institute, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Tan H, Zhang S, Zhang Z, Zhang J, Wang Z, Liao J, Qiu X, Jia E. Neutrophil extracellular traps promote M1 macrophage polarization in gouty inflammation via targeting hexokinase-2. Free Radic Biol Med 2024; 224:540-553. [PMID: 39277122 DOI: 10.1016/j.freeradbiomed.2024.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Peptidylarginine deiminase 4 (PAD4)-dependent neutrophil extracellular trap (NET) formation is a new neutrophil death mechanism. Increased NET formation has been demonstrated to be associated with gouty inflammation. Macrophages release proinflammatory mediators and chemokines in acute gouty inflammation and subsequently lead to inflammatory cascades. However, whether NETs regulate macrophage function and polarization and further contribute to gout development remains unclear. Herein, we investigated the relationship between monosodium urate (MSU) crystal-induced NETs and macrophages and the associated mechanisms in gouty inflammation. Elevated NET formation and CD86+ macrophage infiltration were observed in human gouty arthritis (GA). In vitro, MSU crystal-induced NETs or NET-associated histone H3 treatments modulated nod-like receptor protein 3 (NLRP3) inflammasome activation, M1 polarization, and metabolic changes in macrophages. These effects were eliminated by hexokinase-2 (HK-2) silencing. Moreover, NET formation and inflammation were significantly reduced in PAD4-/- GA mice. Pharmacological inhibition of NET formation with Cl-Amidine or NET degradation with DNase Ⅰ significantly reduced M1 polarization of macrophages and ameliorated inflammation in GA mice. In sum, MSU crystal-induced NETs promote M1 polarization and NLRP3 activation in macrophages via targeting HK-2. Cell-free DNA and histone H3 may be the driving elements behind the NET-induced M1 macrophage polarization, NLRP3 activation, and metabolic changes. Targeting NETs could be a potential therapeutic strategy for gout flare.
Collapse
Affiliation(s)
- Haibo Tan
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Shan Zhang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Zhihao Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Jianyong Zhang
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Ziyu Wang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Junlan Liao
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518033, PR China
| | - Xia Qiu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Department of Rheumatism, Shenzhen, 518033, PR China
| | - Ertao Jia
- The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangdong Second Hospital of Traditional Chinese Medicine, Department of Rheumatism, Guangzhou, 510000, PR China.
| |
Collapse
|
41
|
Zhao Y, Wang L, Zhang X, Zhang L, Wei F, Li S, Li Y. Identification of neutrophil extracellular traps genes as potential biomarkers in psoriasis based on bioinformatics analysis. Sci Rep 2024; 14:23848. [PMID: 39394253 PMCID: PMC11470069 DOI: 10.1038/s41598-024-75069-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 10/01/2024] [Indexed: 10/13/2024] Open
Abstract
The epidermal infiltration of neutrophils is a hallmark of psoriasis (PSO) and its activation leads to the release of neutrophil extracellular traps (NETs). However, the molecular mechanism of NETs-related genes (NETRGs) has not been extensively studied in PSO. To define NETs-related-biomarkers for PSO. The GSE13355 and GSE78097 datasets, and NETRGs gene set were included in this study. The datasets used in this study were all microarray data. The weighted gene co-expression network analysis (WGCNA) and machine learning algorithms were used to mine key genes. Later on, single-gene gene set enrichment analysis (GSEA) and immune infiltration analysis were implemented. Finally, the expression of key genes was verified using quantitative real-time fluorescence PCR (qRT-PCR). A total of 3 key genes (S100A9, CLEC7A, and CXCR4) were derived, and they all had excellent diagnostic performance. The single-gene GSEA enrichment results indicated that the key genes were mainly enriched in the chemokine signaling pathway and humoral immune response in the high-expression group, while focal adhesion was enriched in the low-expression group. The correlation analysis indicated that all key genes were strongly negatively correlated with resting mast cells and TGF-β family member receptor, while they were strongly positively correlated with activated CD4 memory T cells and antigen processing and presentation. Lastly, the experimental results showed that the expression trends of key genes were consistent with public database. In this study, we successfully screened three potential PSO diagnostic genes (S100A9, CLEC7A and CXCR4) that were closely related to NETs, and these findings not only provided new molecular marker candidates for the precise diagnosis of PSO patients, but also revealed possible future therapeutic targets. However, further in-depth research and validation were necessary.
Collapse
Affiliation(s)
- Yike Zhao
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Ling Wang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Xiaoguang Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Lihua Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Feng Wei
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Suyue Li
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Yanling Li
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereology in Hebei Province, Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
42
|
Geng X, Wang DW, Li H. The pivotal role of neutrophil extracellular traps in cardiovascular diseases: Mechanisms and therapeutic implications. Biomed Pharmacother 2024; 179:117289. [PMID: 39151311 DOI: 10.1016/j.biopha.2024.117289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
Cardiovascular diseases (CVDs) continue to pose a significant burden on global health, prominently contributing to morbidity and mortality rates worldwide. Recent years have witnessed an increasing recognition of the intricate involvement of neutrophil extracellular traps (NETs) in the pathology of diverse cardiovascular conditions. This review provides a comprehensive analysis of the multifaceted functions of NETs in cardiovascular diseases, shedding light on the impact on atherosclerosis, myocardial infarction, heart failure, myocarditis, atrial fibrillation, aortic stenosis, and the potential therapeutic avenues targeting NETs.
Collapse
Affiliation(s)
- Xinyu Geng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huihui Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
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: 35] [Impact Index Per Article: 35.0] [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
|
44
|
Xv Y, Feng Y, Lin J. CXCR1 and CXCR2 are potential neutrophil extracellular trap-related treatment targets in ulcerative colitis: insights from Mendelian randomization, colocalization and transcriptomic analysis. Front Immunol 2024; 15:1425363. [PMID: 39328405 PMCID: PMC11424450 DOI: 10.3389/fimmu.2024.1425363] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Objectives There is already substantial evidence indicating that neutrophil extracellular trap (NET) formation contributes to the inflammatory cascade in ulcerative colitis (UC). However, the precise regulatory mechanisms governing this process remain elusive. This study aimed to determine the role of NET-related genes in UC and reveal possible mechanisms. Methods Employing a two-sample MR methodology, we investigated the correlations between NET-associated genes (NRGs) and UC with summary data derived from a genome-wide association study (12,366 cases vs. 33,609 controls) and FinnGen (8,279 cases vs. 261,098 controls). The main analysis employed the inverse variance weighted method, supplemented by the MR-Egger method and weighted median method. Sensitivity analysis was conducted to rule out the interference of heterogeneity and pleiotropy among utilized instrument variables. The colocalization analysis was used to determine whether the identified NRGs and UC shared casual variants. Cross-tissue expression analysis was performed to characterize the expression patterns of target NRGs, while multi-gene correlation analysis and GSEA analysis were conducted to explore the mechanisms by which target NRGs promote UC and NET formation. Immunohistochemistry was used to validate the protein expression of target NRGs in the colon tissue of UC patients. Results After the validation of two datasets, seven NRGs were associated with the risk of UC. The higher expression of ITGB2 was associated with increased UC risk, while the expression of CXCR1, CXCR2, IRAK4, MAPK3, SIGLEC14, and SLC22A4 were inversely associated with UC risk. Colocalization analysis supported the correlation between CXCR1/2 and UC risk. Expression analysis indicated that CXCR1/2 were down-regulated in peripheral blood, but up-regulated in colon tissue. GSEA analysis and correlation analysis indicated that CXCR1/2 promoted UC and NET formation through neutrophil chemotaxis and PAD4-mediated pathways, separately. Immunohistochemical results confirmed the high expression of CXCR1/2 in colon tissues of UC patients. Conclusions Our study identified CXCR1/2 as candidate targets in UC among all NRGs through multi-method argumentation, providing new insights of the regulation mechanisms of NET formation in the pathogenesis of UC.
Collapse
Affiliation(s)
- Yichuan Xv
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyi Feng
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Macáková K, Borbélyová V, Tekeľová M, Janko J, Pastorek M, Hokša R, Moravanský N, Šteňová E, Vlková B, Celec P. Effects of exogenous deoxyribonuclease I in collagen antibody-induced arthritis. J Inflamm (Lond) 2024; 21:36. [PMID: 39251994 PMCID: PMC11386490 DOI: 10.1186/s12950-024-00403-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is associated with a high concentration of extracellular DNA (ecDNA). This could be a consequence of the inflammation, but the ecDNA could also be involved in the unknown etiopathogenesis of RA. Clearance of ecDNA is hypothesized to prevent the development of RA. This study aimed to analyze the effects of exogenous deoxyribonuclease I (DNase I) administration in an animal model of RA. METHODS The collagen antibody-induced arthritis (CAIA) model of RA was induced in adult female DBA/1J mice. CAIA mice were treated with saline or DNase I (10 mg/kg) every 12 h for the whole duration of the experiment. Arthritic scores were assessed. Paw volume and temperature were assessed using a plethysmometer and a thermal camera, respectively. Plasma ecDNA and its subcellular origin were analyzed using fluorometry and real-time PCR. DNase activity was quantified with single radial enzyme diffusion method. RESULTS The CAIA model was successfully induced as proved by a higher volume, temperature and the overall arthritis score in comparison to controls. The administration of DNase I resulted in a nearly two-fold increase in serum DNase activity. Still, it did affect neither plasma ecDNA, nor the arthritis score or other measures of joint inflammation. CONCLUSION Our results suggest that exogenous DNase I does not prevent the development of CAIA in mice. Whether this is true for other animal models of arthritis or clinical RA requires further research. EcDNA does not seem to be involved in the pathogenesis of CAIA. Additional studies are also needed to elucidate the role of ecDNA in the development of RA, focusing especially on its origin and inhibition of ecDNA release.
Collapse
Affiliation(s)
- Kristína Macáková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Mária Tekeľová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Jakub Janko
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Michal Pastorek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Richard Hokša
- Health Care Surveillance Authority Department, Department of Pathology, Antolská 11, 851 07 Bratislava, Slovakia
- Forensic.sk Institute of Forensic Medical Expertise, Boženy Němcovej 8, 811 01 Bratislava, Slovakia
| | - Norbert Moravanský
- Institute of Forensic Medicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Forensic.sk Institute of Forensic Medical Expertise, Boženy Němcovej 8, 811 01 Bratislava, Slovakia
| | - Emöke Šteňová
- 1st Department of Internal Medicine, Faculty of Medicine, University Hospital, Comenius University, Mickiewiczova 13, 813 69 Bratislava, Slovakia
| | - Barbora Vlková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, Bratislava, 811 08, Slovakia.
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia.
| |
Collapse
|
46
|
Quan M, Zhang H, Han X, Ba Y, Cui X, Bi Y, Yi L, Li B. Single-Cell RNA Sequencing Reveals Transcriptional Landscape of Neutrophils and Highlights the Role of TREM-1 in EAE. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200278. [PMID: 38954781 PMCID: PMC11221915 DOI: 10.1212/nxi.0000000000200278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 05/06/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND AND OBJECTIVES Neutrophils, underestimated in multiple sclerosis (MS), are gaining increased attention for their significant functions in patients with MS and the experimental autoimmune encephalomyelitis (EAE) animal model. However, the precise role of neutrophils in cervical lymph nodes (CLNs), the primary CNS-draining lymph nodes where the autoimmune response is initiated during the progression of EAE, remains poorly understood. METHODS Applying single-cell RNA sequencing (scRNA-seq), we constructed a comprehensive immune cell atlas of CLNs during development of EAE. Through this atlas, we concentrated on and uncovered the transcriptional landscape, phenotypic and functional heterogeneity of neutrophils, and their crosstalk with immune cells within CLNs in the neuroinflammatory processes in EAE. RESULTS Notably, we observed a substantial increase in the neutrophil population in EAE mice, with a particular emphasis on the significant rise within the CLNs. Neutrophils in CLNs were categorized into 3 subtypes, and we explored the specific roles and developmental trajectories of each distinct neutrophil subtype. Neutrophils were found to engage in extensive interactions with other immune cells, playing crucial roles in T-cell activation. Moreover, our findings highlighted the strong migratory ability of neutrophils to CLNs, partly regulated by triggering the receptor expressed on myeloid cells 1 (TREM-1). Inhibiting TREM1 with LR12 prevents neutrophil migration both in vivo and in vitro. In addition, in patients with MS, we confirmed an increase in peripheral neutrophils with an upregulation of TREM-1. DISCUSSION Our research provides a comprehensive and precise single-cell atlas of CLNs in EAE, highlighting the role of neutrophils in regulating the periphery immune response. In addition, TREM-1 emerged as an essential regulator of neutrophil migration to CLNs, holding promise as a potential therapeutic target in MS.
Collapse
Affiliation(s)
- Moyuan Quan
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Huining Zhang
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Xianxian Han
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Yongbing Ba
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Xiaoyang Cui
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Yanwei Bi
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Le Yi
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| | - Bin Li
- From the Department of Neurology (M.Q., H.Z., L.Y., B.L.), The Second Hospital of Hebei Medical University; the Key Laboratory of Hebei Neurology, Hebei Medical University, Ministry of Education, (M.Q., H.Z., L.Y., B.L.); and the Key Laboratory of Neurology of Hebei Province, (M.Q., H.Z., L.Y., B.L.), Shijiazhuang, Hebei, China; Department of Neurology (X.H.), Zhongshan People's Hospital, China; OE Biotech Co. (Yongbing Ba), Ltd. Shanghai, China; and School of Basic Medicine (X.C., Yanwei Bi), Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
47
|
Li Z, Lu Q. The role of neutrophils in autoimmune diseases. Clin Immunol 2024; 266:110334. [PMID: 39098706 DOI: 10.1016/j.clim.2024.110334] [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: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
Historically, neutrophils have been primarily regarded as short-lived immune cells that act as initial responders to antibacterial immunity by swiftly neutralizing pathogens and facilitating the activation of adaptive immunity. However, recent evidence indicates that their roles are considerably more complex than previously recognized. Neutrophils comprise distinct subpopulations and can interact with various immune cells, release granular proteins, and form neutrophil extracellular traps. These functions are increasingly recognized as contributing factors to tissue damage in autoimmune diseases. This review comprehensively examines the physiological functions and heterogeneity of neutrophils, their interactions with other immune cells, and their significance in autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, antiphospholipid syndrome, antineutrophil cytoplasmic antibody-associated vasculitis, multiple sclerosis, and others. This review aims to provide a deeper understanding of the function of neutrophils in the development and progression of autoimmune disorders.
Collapse
Affiliation(s)
- Zhuoshu Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences &Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences &Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
48
|
Man SM, Kanneganti TD. Innate immune sensing of cell death in disease and therapeutics. Nat Cell Biol 2024; 26:1420-1433. [PMID: 39223376 DOI: 10.1038/s41556-024-01491-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Innate immunity, cell death and inflammation underpin many aspects of health and disease. Upon sensing pathogens, pathogen-associated molecular patterns or damage-associated molecular patterns, the innate immune system activates lytic, inflammatory cell death, such as pyroptosis and PANoptosis. These genetically defined, regulated cell death pathways not only contribute to the host defence against infectious disease, but also promote pathological manifestations leading to cancer and inflammatory diseases. Our understanding of the underlying mechanisms has grown rapidly in recent years. However, how dying cells, cell corpses and their liberated cytokines, chemokines and inflammatory signalling molecules are further sensed by innate immune cells, and their contribution to further amplify inflammation, trigger antigen presentation and activate adaptive immunity, is less clear. Here, we discuss how pattern-recognition and PANoptosome sensors in innate immune cells recognize and respond to cell-death signatures. We also highlight molecular targets of the innate immune response for potential therapeutic development.
Collapse
Affiliation(s)
- Si Ming Man
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.
| | | |
Collapse
|
49
|
Chen J, Cao Y, Xiao J, Hong Y, Zhu Y. The emerging role of neutrophil extracellular traps in the progression of rheumatoid arthritis. Front Immunol 2024; 15:1438272. [PMID: 39221253 PMCID: PMC11361965 DOI: 10.3389/fimmu.2024.1438272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. Neutrophil extracellular traps (NETs are NETwork protein structures activated by neutrophils to induce the cleavage and release of DNA-protein complexes). Current studies have shown the critical involvement of NETs in the progression of autoimmune diseases, Neutrophils mostly gather in the inflammatory sites of patients and participate in the pathogenesis of autoimmune diseases in various ways. NETs, as the activated state of neutrophils, have attracted much attention in immune diseases. Many molecules released in NETs are targeted autoantigens in autoimmune diseases, such as histones, citrulline peptides, and myeloperoxidase. All of these suggest that NETs have a direct causal relationship between the production of autoantigens and autoimmune diseases. For RA in particular, as a disorder of the innate and adaptive immune response, the pathogenesis of RA is inseparable from the generation of RA. In this article, we investigate the emerging role of NETs in the pathogenesis of RA and suggest that NETs may be an important target for the treatment of inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Jingjing Chen
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yang Cao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Jing Xiao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yujie Hong
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yan Zhu
- The Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
50
|
Guan X, Guan X, Zhao Z, Yan H. NETs: Important players in cancer progression and therapeutic resistance. Exp Cell Res 2024; 441:114191. [PMID: 39094902 DOI: 10.1016/j.yexcr.2024.114191] [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: 04/21/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Neutrophil extracellular traps (NETs) are web-like structures composed of cytoplasmic contents, DNA chromatin and various granular proteins released by neutrophils in response to viruses, bacteria, immune complexes and cytokines. Studies have shown that NETs can promote the occurrence, development and metastasis of tumors. In this paper, the mechanism underlying the formation and degradation of NETs and the malignant biological behaviors of NETs, such as the promotion of tumor cell proliferation, epithelial mesenchymal transition, extracellular matrix remodeling, angiogenesis, immune evasion and tumor-related thrombosis, are described in detail. NETs are being increasingly studied as therapeutic targets for tumors. We have summarized strategies for targeting NETs or interfering with NET-cancer cell interactions and explored the potential application value of NETs as biomarkers in cancer diagnosis and treatment, as well as the relationship between NETs and therapeutic resistance.
Collapse
Affiliation(s)
- Xiaoying Guan
- Pathology Department, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Xiaoli Guan
- General Medicine Department, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Zhiqiang Zhao
- Pathology Department, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China
| | - Hong Yan
- Pathology Department, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, Gansu, China.
| |
Collapse
|