1
|
Deng Z, Mei S, Ouyang Z, Wang R, Wang L, Zou B, Dai J, Mao K, Li Q, Guo Q, Yi C, Meng F, Xie M, Zhang X, Wang R, Deng T, Wang Z, Li X, Wang Q, Liu B, Tian X. Dysregulation of gut microbiota stimulates NETs-driven HCC intrahepatic metastasis: therapeutic implications of healthy faecal microbiota transplantation. Gut Microbes 2025; 17:2476561. [PMID: 40099491 PMCID: PMC11925110 DOI: 10.1080/19490976.2025.2476561] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/04/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
The stringent regulation of intrahepatic metastases is essential for improving survival outcomes in patients with hepatocellular carcinoma (HCC). This study investigated the impact of gut microbiota on intrahepatic metastasis of HCC and evaluated the therapeutic potential of healthy fecal microbiota transplantation (FMT). Dysregulation of the gut microbiota, characterized by a significant reduction in the abundance of beneficial bacteria, such as Anaerotruncus colihominis and Dysosmobacter welbionis, was observed in patients with intrahepatic metastatic HCC. A human flora-associated (HFA) intrahepatic metastatic HCC mouse model was successfully established through consecutive 4 weeks of human-mouse FMT. Dysregulation of gut microbiota promoted intrahepatic metastasis in the mouse model, primarily by enhancing neutrophil-mediated inflammatory responses and lead to excessive formation of neutrophil extracellular traps (NETs). Consequently, it promoted tumor vascular growth and tissue necrosis, resulting in intrahepatic metastasis of HCC. Notably, FMT from healthy donors mitigated these pathological processes. This study elucidated the role and mechanism of dysregulated gut microbiota in promoting intrahepatic metastasis of HCC. Healthy FMT emerges as a promising novel therapeutic strategy for preventing and treating intrahepatic metastasis of HCC.
Collapse
Affiliation(s)
- Zhe Deng
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Si Mei
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Changsha, Hunan, China
- Department of Physiology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhaoguang Ouyang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Ruoyu Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lihuai Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Bo Zou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingjing Dai
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Kexin Mao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qian Li
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qianqian Guo
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chun Yi
- Department of Pathology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fanying Meng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mingxia Xie
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xue Zhang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rongrong Wang
- Hunan Province Integrated Traditional Chinese and Western Medicine Hospital, Changsha, Hunan, China
| | - Tianhao Deng
- Hunan Province Integrated Traditional Chinese and Western Medicine Hospital, Changsha, Hunan, China
| | - Zhenyu Wang
- JCY Biotech Ltd., Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiaozheng Li
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, China
| | - Qing Wang
- Shanghai OE Biotech Co. Ltd, Shanghai, China
| | - Bin Liu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Changsha, Hunan, China
| |
Collapse
|
2
|
Givian A, Azizan A, Jamshidi A, Mahmoudi M, Farhadi E. Iron metabolism in rheumatic diseases. J Transl Autoimmun 2025; 10:100267. [PMID: 39867458 PMCID: PMC11763848 DOI: 10.1016/j.jtauto.2025.100267] [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: 11/09/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Iron is a crucial element for living organism in terms of oxygen transport, hematopoiesis, enzymatic activity, mitochondrial respiratory chain function and also immune system function. The human being has evolved a mechanism to regulate body iron. In some rheumatic diseases such as rheumatoid arthritis (RA), systemic lupus erythematous (SLE), systemic sclerosis (SSc), ankylosing spondylitis (AS), and gout, this balanced iron regulation is impaired. Altered iron homeostasis can contribute to disease progression through ROS production, fibrosis, inflammation, abnormal bone homeostasis, NETosis and cell senescence. In this review, we have focused on the iron metabolism in rheumatic disease and its role in disease progression.
Collapse
Affiliation(s)
- Aliakbar Givian
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Amin Azizan
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Shen J, Lin H, Mo K, Liang Z, Zhang Y, Quan H, Wang X, Zhang C, Chen C. Bidirectional roles of neutrophil extracellular traps in oral microbiota carcinogenesis: A systematic review. Transl Oncol 2025; 56:102361. [PMID: 40239243 PMCID: PMC12022684 DOI: 10.1016/j.tranon.2025.102361] [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: 08/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) are network structures composed of DNA, histones, and antimicrobial proteins,released by activated neutrophils to trap and eliminate extracellular pathogens. Recent research has demonstrated a strong correlation between NETs and various diseases, including immune dysregulation, thrombosis, and malignancies. This review synthesizes current research on NETs, focusing on its biological role in oral squamous cell carcinoma (OSCC) and explores its potential in treating. METHODS A literature review in the PubMed database was conducted to examine the impact of NETs on the homeostasis of oral microbiota and the involvement in the development of oral microbiota-related carcinogenesis. RESULTS Various microorganisms, including Porphyromonas gingivalis, Fusobacterium nucleatum, Streptococcus spp., along with Candida albicans, as well as certain viruses such as Human papillomavirus (HPV), Human herpes virus 8 (HHV-8), and Herpes simplex virus-1 (HSV-1)are regulated by NETs during oral colonization and proliferation and have been identified as contributors to the pathogenesis of oral squamous cell carcinoma. NETs have been shown to play a dual role in the carcinogenic process of oral microbiota in humans. At the initial stage of tumor formation, NETs inhibit tumorigenesis by eliminating tumorigenic bacteria that infiltrated the tumor; however, following tumor establishment, various cytokines and chemokines that promote tumor progression are released by neutrophils during the NETs formation. CONCLUSION This article reviews the oncogenic mechanisms of NETs in the oral microbiota, with potential implications for early tumor detection and the development of microbe-targeted therapies.
Collapse
Affiliation(s)
- Jie Shen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Haitao Lin
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Kangnan Mo
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Zhong Liang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yan Zhang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huatao Quan
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Xing Wang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chenping Zhang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Chao Chen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
4
|
Majeed NS, Mohammed MH, Hatem ZA, El-Sehrawy AAMA, Ganesan S, Singh A, Akoul MA, Sudan P, Singh R, Hamad HA. Interplay between NETosis and the lncRNA-microRNA regulatory axis in the immunopathogenesis of cancer. J Physiol Biochem 2025:10.1007/s13105-025-01082-x. [PMID: 40358898 DOI: 10.1007/s13105-025-01082-x] [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: 11/20/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Abstract
Neutrophil extracellular traps (NETs), web-like complex structures secreted by neutrophils, have emerged as key players in the modulation of immune responses and the immunopathogenesis of immune disorders. Initially described for their antimicrobial function, NETs now play a part in the fundamental processes of cancer biology, including cancer initiation, metastatic dissemination, and immune evasion strategies. NETs hijack anti-tumor immunity by entrapping circulating cancer cells, fostering the growth of tumors, and reorganizing the tumor microenvironment such that it is pro-malignancy. Emerging evidence emphasizes the role of NETosis coupled with non-coding RNAs-long non-coding RNAs (lncRNAs) and microRNAs (miRNAs)-as key regulators of gene expression and controllers of processes vital for cancer growth, such as immune response and programmed cell death processes like apoptosis, necroptosis, pyroptosis, and ferroptosis. Aberrantly expressed non-coding RNAs have been attributed to immune dysregulation and excessive NET production, promoting tumor growth. NETs are also associated with a myriad of pathological conditions, such as autoimmune disorders, cystic fibrosis, sepsis, and thrombotic disorders. New therapeutic approaches-such as DNase therapy and PAD4 inhibitors-target NET production and their degradation to modify immune function and the efficiency of immunotherapies. Further clarification of the intricate interactions of NETosis, lncRNAs, and miRNAs has the potential to establish new strategies for the suppression of the growth of tumors and preventing immune evasion. This review seeks to elucidate the interactions between NETosis and the regulatory networks involving non-coding RNAs that significantly contribute to the immunopathogenesis of cancer.
Collapse
Affiliation(s)
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques department, College of Health and medical technology, Al-Maarif University, Anbar, Iraq.
| | - Zainab Amer Hatem
- College of Science, Biotechnology Department, Diyala University, Diyala, Iraq
| | | | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Marwa Azeez Akoul
- Biotechnology Department, College of Applied Science, Fallujah University, Anbar, Iraq
| | - Puneet Sudan
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Roshni Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Hamad Ali Hamad
- Department of Pathological Analysis, Collage of Applied Sciences, University of Fallujah, Fallujah, Iraq
| |
Collapse
|
5
|
Blair KM, Bohinc DJ, Bane KL, Warnock M, Abuaita B, Gura C, Grinsztejn E, Marshall SH, Wilson BM, Bonomo RA, Tambralli A, Knight JS, O'Riordan MX, Lawrence DA, Stavrou EX, Sandkvist M. Acinetobacter Baumannii Secreted Protease CpaA Inhibits Factor XII-Mediated Bradykinin Generation and Neutrophil Activation. Circ Res 2025. [PMID: 40357548 DOI: 10.1161/circresaha.124.324764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND FXII (coagulation factor XII) is best known for its roles in the contact and kallikrein-kinin pathways. FXII is converted to its active enzyme (FXIIa [activated factor XII]) by PKa (plasma kallikrein) or its unique ability to autoactivate on bacterial or other biologic surfaces. In vivo, FXIIa initiates the intrinsic coagulation pathway and promotes inflammation by reciprocal activation of prekallikrein, which cleaves HK (high-molecular-weight kininogen) to liberate bradykinin. CpaA (coagulation targeting metallo-endopeptidase of A baumannii) is a secreted metalloprotease identified in a human clinical isolate of Acinetobacter baumannii that cleaves FXII at O-linked glycosylated sites, inhibiting contact activation. While CpaA facilitates a modest in vivo fitness advantage in mice, the role of CpaA in human infection remains unclear. As such, the objectives of this study were to characterize the structural details of the interaction between CpaA, FXII, and the KKSs (kallikrein-kinin systems) and to determine the downstream consequences on thromboinflammatory responses. METHODS The effect of purified CpaA on the coagulant activity of FXII and the generation of bradykinin was characterized. Neutrophil signaling, flow cytometry, and functional assays were performed to define how CpaA-mediated cleavage of FXII affects innate immune functions. Bacterial killing by human neutrophils was performed with wild-type and mutant A baumannii strains lacking CpaA. RESULTS We found that CpaA cleaves both FXII zymogen and FXIIa but not beta Factor XII. However, cleavage of FXIIa by CpaA does not significantly inhibit its clotting activity, demonstrating that CpaA does not inactivate FXIIa, but rather prevents activation of zymogen FXII. CpaA also cleaves HK, resulting in reduced kallikrein activation and bradykinin generation. We previously identified that zymogen FXII interacts with the urokinase receptor on neutrophils and upregulates neutrophil activation. Here, we demonstrate that CpaA cleaves neutrophil FXII, resulting in reduced Akt2 phosphorylation, chemotaxis, oxidative burst, and neutrophil extracellular trap formation. Importantly, CpaA decreases the human neutrophil killing efficiency of A baumannii in culture. CONCLUSIONS These data identify a role for FXII in responding to bacterial infection and suggest that by inhibiting the contact and KKSs and impairing neutrophil activation, CpaA may blunt the innate immune response and help prevent the elimination of A baumannii from the human host.
Collapse
Affiliation(s)
- Kris M Blair
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Now with Fred Hutch/University of Washington/Seattle Children's Cancer Consortium (K.M.B.)
| | - Dillon J Bohinc
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Kara L Bane
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Mark Warnock
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Basel Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge (B.A.)
| | - Colby Gura
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Eduarda Grinsztejn
- Hematology and Oncology Division, Department of Medicine, University Hospitals Cleveland Medical Center, OH (E.G.)
| | - Steven H Marshall
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Brigid M Wilson
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Robert A Bonomo
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
- Clinician Scientist Investigator, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH.(R.A.B.)
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), OH (R.A.B.)
| | - Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Evi X Stavrou
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
- Medicine Service, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH (E.X.S.)
| | - Maria Sandkvist
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| |
Collapse
|
6
|
Hu H, Peng Y, Wang CC, Chen J, Yu X, Chen X, Ouyang H, Huang Q, Ma J, Yin Q, Ma L, Ding Z, Zhang M, Ren H, Zheng J, Chen W, Tao Z, Liu R, Chen L, Wang X, Zhang T, Zhong M. Neutrophil extracellular traps induce trophoblasts pyroptosis via enhancing NLRP3 lactylation in SLE pregnancies. J Autoimmun 2025; 153:103411. [PMID: 40179478 DOI: 10.1016/j.jaut.2025.103411] [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: 01/11/2025] [Revised: 02/12/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder primarily affecting women during the reproductive years, often complicating pregnancy outcomes with elevated levels of neutrophil extracellular traps (NETs) infiltration. However, potential impacts of NETs on placental trophoblasts in SLE and the underlying molecular mechanisms remain unclear. To address this, transcriptome sequencing was conducted on placentas collected from seven pregnant women with SLE and six healthy pregnant controls to identify SLE-specific placental features. The effects of NETs were further assessed in MRL/lpr lupus-prone mice and pristane-induced lupus (PIL) mice, focusing on pregnancy outcomes and placental pathology. In vitro, trophoblasts were stimulated with NETs derived from patients with SLE, followed by molecular analyses such as transcriptomic, cellular energy metabolism assays and liquid chromatography-tandem mass spectrometry to explore the effects and mechanisms of NETs. Results showed elevated NETs were observed in the placentas of both patients with SLE and lupus mouse models, accompanied by activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. Treatment with DNase I significantly improved pregnancy outcomes in MRL/lpr mice, while the use of peptidyl arginine deiminase 4 (PAD4)-deficient mice was beneficial on the pregnancy outcomes of PIL mice. Furthermore, SLE-derived NETs activated pyroptosis in trophoblasts by promoting glycolysis and subsequent lactylation of NLRP3. These findings highlight that NETs contribute to placental damage in SLE by inducing the lactylation of the NLRP3 inflammasome in trophoblasts, demonstrating the therapeutic potential of inhibiting NETs to improve placental function.
Collapse
Affiliation(s)
- Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Obstetrics and Gynaecology, The Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - You Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Obstetrics and Gynaecology, The Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jun Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Obstetrics and Gynaecology, The Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Haotong Ouyang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Qin Huang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Ma
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lien Ma
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziling Ding
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Minyi Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Ren
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaman Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenqian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zixin Tao
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ruiyan Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Chen
- Department of Obstetrics and Gynaecology, The Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Xuefei Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Tao Zhang
- Department of Obstetrics and Gynaecology, The Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Sino-US Center of Translational Medicine for Development Disabilities, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Huang S, Zhan Z, Xu F, Liu X, Fang Z, Wu W, Liang Z, Liu G, Wang M, da Silva HS, Luo X, Mo K. Ganoderma lucidum spore powder alleviates rheumatoid arthritis-associated pain hypersensitivity through inhibiting accumulation, N1 polarization, and ROS production of neutrophils in mice. Front Immunol 2025; 16:1569295. [PMID: 40370462 PMCID: PMC12075414 DOI: 10.3389/fimmu.2025.1569295] [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: 01/31/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic condition characterized by joint pain that significantly impairs patients' work and daily lives. The limited understanding of the pathological mechanisms underlying RA-related pain poses challenges for effective clinical pain management. Ganoderma lucidum spore powder (GLSP) has demonstrated therapeutic benefits in various diseases, with no reported toxicity or adverse effects. Methods This study investigates the role of neutrophils in the pathological mechanisms of RA-related pain using collagen-induced arthritis (CIA) mice and an ex vivo neutrophil model. A combination of techniques, including animal models, flow cytometry, behavioral testing, cell adoptive transfer, and network pharmacology analysis, was employed to evaluate the effects and targets of GLSP on pain symptoms and neutrophil activity in CIA mice. Results Flow cytometric analysis revealed the accumulation and activation of neutrophils in the paws of CIA mice. Furthermore, the levels of pro-inflammatory CD95+ neutrophil subpopulations (N1 state) and ROS+ cells in the affected paws were positively correlated with the severity of mechanical allodynia and heat hyperalgesia observed in these mice. Our findings indicate that oral administration of GLSP significantly alleviates joint destruction, paw swelling, and pain hypersensitivity in CIA mice. Notably, GLSP reversed CIA-induced neutrophil accumulation, N1 polarization, and reactive oxygen species (ROS) production. Both network pharmacology target prediction and in vivo/in vitro experimental validation indicated that GLSP inhibits N1 polarization and ROS production in neutrophils by modulating the TNF-α signaling pathway, thus exerting RA-specific analgesic effects. Discussion In summary, this study offers new insights into the pathological mechanisms of RA-related pain and demonstrates that neutrophil accumulation, N1 polarization, and ROS production contribute to RA-related pain. GLSP alleviates RA-related pain by inhibiting the pro-inflammatory phenotype of neutrophils, highlighting its potential for clinical translation in the treatment of RA.
Collapse
Affiliation(s)
- Sen Huang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhaochun Zhan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Fei Xu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaolin Liu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenning Fang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenbo Wu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhile Liang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guoguo Liu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mengyuan Wang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Helena Soares da Silva
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Luo
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Shunde Hospital, Southern Medical University, Foshan, China
- Institute of Perioperative Medicine and Organ Protection, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Mo
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Perioperative Medicine and Organ Protection, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Jesus Gonzalez-Contreras FD, Gutierrez-Vidal RG, Zarate X. A recombinant human SLPI variant suppresses the formation of neutrophil extracellular traps at low concentrations in vitro. Protein Expr Purif 2025; 232:106721. [PMID: 40280458 DOI: 10.1016/j.pep.2025.106721] [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: 02/14/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Neutrophil extracellular traps (NETs) are web-like structures released by neutrophils to trap and kill microbes. While NETs are crucial in host defense, excessive formation can lead to autoimmune diseases. Currently, no specific drugs target NETs directly; however, some medications can indirectly modulate their formation. The secretory leukocyte protease inhibitor (SLPI) is a small protein that inhibits the activity of neutrophil elastase (NE), an enzyme essential for NETs formation. By inhibiting NE activity, SLPI prevents the chromatin from decondensing, which is necessary for NETs to form; this suggests that SLPI may protect by preventing excessive NETs development. However, evidence indicates that NE can inactivate SLPI by cleaving its N-terminus. This action can create a protease/antiprotease imbalance, potentially leading to detrimental consequences for the host. In this study, we produced a recombinant variant of SLPI (SLPI-S15G-A16G: rSLPIv) using recombinant DNA technology, expressed in Escherichia coli SHuffle T7. The protein was tagged with the small metal-binding protein (SmbP) to facilitate its expression and purification through immobilized metal-affinity chromatography. Our results demonstrated that rSLPIv exhibited immunomodulatory activity at a concentration of 10 nM in neutrophils that had been prestimulated with PMA. It reduced NETs formation by 30 % and maintained this effect for up to 6 h. Confocal microscopy confirmed these findings, revealing a rSLPIv-dependent reduction in neutrophil nuclear expansion. Thus, rSLPIv shows significant suppressive activity on NETs formation at low concentrations, making it a potential candidate as an immunotherapeutic agent.
Collapse
Affiliation(s)
- Felipe de Jesus Gonzalez-Contreras
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, Ciudad Universitaria, San Nicolas de los Garza, NL, 66455, Mexico
| | - Roxana Guadalupe Gutierrez-Vidal
- Programa de Investigadoras e Investigadores por México CONAHCYT, Mexico; Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, Unidad Monterrey, Via del Conocimiento 201, Parque de Investigacion e Innovacion Tecnologica, Apodaca, NL, 66600, Mexico
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, Ciudad Universitaria, San Nicolas de los Garza, NL, 66455, Mexico.
| |
Collapse
|
9
|
Wang J, Liang Y, Meng Y, Chen J, Fang L, Yang H, Li P. Assessment of lncRNA biomarkers based on NETs for prognosis and therapeutic response in ovarian cancer. Sci Rep 2025; 15:13042. [PMID: 40234525 PMCID: PMC12000398 DOI: 10.1038/s41598-025-97548-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 04/04/2025] [Indexed: 04/17/2025] Open
Abstract
Ovarian cancer (OC) usually progresses rapidly and is associated with high mortality, while a reliable clinical factor for OC patients to predict prognosis is currently lacking. Recently, the pathogenic role of neutrophils releasing neutrophil extracellular traps (NETs) in various cancers including OC has gradually been recognized. The study objective was to determine whether NETs-related biomarkers can be used to accurately predict the prognosis and guide clinical decision-making in OC. In this study, we utilized univariate and multivariate Cox regression to identify key prognostic features and developed a model with six NETs-related lncRNAs, selected via LASSO regression. The model's predictive capability was assessed through Kaplan-Meier, ROC, and Cox analyses. To understand the model's mechanisms, we conducted GO term analysis, KEGG pathway enrichment, and GSEA. We also analyzed gene mutation status, tumor mutation load, survival rates, and model correlation. Additionally, we compared immune functions, immune checkpoint expression, and chemotherapy sensitivity between risk groups. Besides, we validated the model's predictive value using test data and tissues acquired from our institution. Finally, we performed in vitro and in vivo experiments to confirm the expression of model lncRNAs and the cellular level function of GAS5. We developed a model using six NETs-associated lncRNAs: GAS5, GBP1P1, LINC00702, LINC01933, LINC02362, and ZNF687-AS1. The model's predictive performance, evaluated via ROC curve, was compared with traditional clinicopathological features. GO process analysis highlighted molecular functions related to antigen binding and immune system biological processes. Variations were observed in transcription regulators affecting immune response, inflammation, cytotoxicity, and regulation. We also predicted IC50 values for chemotherapeutic drugs (bexarotene, bicalutamide, embelin, GDC0941, and thapsigargin) in high- and low-risk groups, finding higher IC50 values in low-risk patients. The risk model's robustness was validated using OC cells, tissues, and clinical datas.
Collapse
Affiliation(s)
- Jingmeng Wang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Yusen Liang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Yimei Meng
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Jialin Chen
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Lei Fang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huike Yang
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China.
| | - Peiling Li
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
10
|
Mak KM, Shekhar AC, Ding SY. Neutrophil extracellular traps mediate pathophysiology of hepatic cells during liver injury. Anat Rec (Hoboken) 2025. [PMID: 40219700 DOI: 10.1002/ar.25673] [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: 02/07/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Neutrophil extracellular traps (NETs) are web-like, bactericidal structures produced by neutrophils and are composed principally of extracellular DNA, histones, elastase, and myeloperoxidase, among other components. NET formation is an innate immune response that is beneficial for pathogen killing and clearance. However, excessive NET formation and clearance defects can lead to inflammation and induce damage to host organs. NETs are also implicated in the development of noninfectious inflammatory disorders, such as liver injury in chronic liver diseases. The liver parenchyma contains hepatocytes, liver sinusoidal endothelial cells, Kupffer cells, and hepatic stellate cells. Each of these cells possesses unique structures and functions, and their interactions with NETs result in pathophysiological changes contributing to liver injury. This review updates the findings related to the modes of action and molecular mechanisms by which NETs modulate the pathophysiology of various hepatic cells and potentiate liver injury. The article also reviews the roles of NETs in hepatic ischemia reperfusion injury, hepatocellular carcinoma pathogenesis, and cancer metastasis. Last, we examine data to determine whether NETs induce crosstalk among various hepatic cells during liver injury and to identify future research directions.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya C Shekhar
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Selena Y Ding
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
11
|
Li N, Deshmukh MV, Sahin F, Hafza N, Ammanath AV, Ehnert S, Nüssler A, Weber ANR, Jin T, Götz F. Staphylococcus aureus thermonuclease NucA is a key virulence factor in septic arthritis. Commun Biol 2025; 8:598. [PMID: 40210969 PMCID: PMC11986129 DOI: 10.1038/s42003-025-07920-4] [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: 08/02/2024] [Accepted: 03/11/2025] [Indexed: 04/12/2025] Open
Abstract
Septic arthritis, primarily caused by Staphylococcus aureus, poses a significant risk of both mortality and morbidity due to its aggressive nature. The nuc1-encoded thermonuclease NucA of S. aureus degrades extracellular DNA/RNA, allowing the pathogen to escape neutrophil extracellular traps (NETs) and maintain the infection unabated. Here we show that in the mouse model for hematogenous septic arthritis, the Δnuc1 mutant is much less pathogenic and the severity of clinical septic arthritis is markedly reduced, including decreased weight loss, lower kidney bacterial load, reduced bone erosion, and much less IL-6 production. In vitro, S. aureus genomic DNA induces a robust TNF-α response in macrophage-like RAW 264.7 cells abrogated when the DNA is degraded by NucA. Moreover, the wild type induces high levels of TNF-α, IL-10, and IL-6 in neutrophils and osteoblast-like SAOS-2 cells, respectively. NucA exacerbates septic arthritis by increasing extracellular and intracellular survival of bacteria.
Collapse
Affiliation(s)
- Ningna Li
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Meghshree Vinod Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Filiz Sahin
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Nourhane Hafza
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | | | - Sabrina Ehnert
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Andreas Nüssler
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, Section Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Cani E, Dwivedi DJ, Carlin S, Sharma N, Chen A, Liaw PC. Impact of peptidylarginine deiminase 4 (PAD4) deficiency in a fecal-induced peritonitis model of sepsis. J Thromb Haemost 2025:S1538-7836(25)00205-3. [PMID: 40199446 DOI: 10.1016/j.jtha.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Peptidylarginine deiminase 4 (PAD4) citrullinates histones, enabling the release of neutrophil extracellular traps. While neutrophil extracellular traps capture and kill pathogens, they also drive immunothrombosis, potentially worsening sepsis outcomes. However, it remains unclear whether PAD4 deficiency is beneficial or harmful in sepsis. OBJECTIVES To evaluate the impact of PAD4 deficiency in a fecal-induced peritonitis sepsis model, with and without antibiotic treatment, and incorporating fluid resuscitation in both sexes. METHODS Wild-type and PAD4 knockout (PAD4-/-) C57Bl/6 mice received intraperitoneal injections of fecal slurry (0.6 mg/g). Mice received buprenorphine every 8 hours and antibiotics/fluids every 12 hours. Survival studies were also conducted without antibiotics at a reduced fecal dose (0.4 mg/g). Mice were culled at 8 hours or 48 hours after infection. Organs, blood, and peritoneal cavity fluid were collected. Plasma levels of interleukin (IL)-6, IL-10, cell-free DNA, and thrombin-antithrombin were quantified, as well as bacterial loads in blood and peritoneal cavity fluid. Organ histology/immunohistochemistry was performed. RESULTS Female PAD4-/- mice had worsened survival compared with female wild-type mice. Male mice exhibited worse survival than females in both strains. Antibiotics eliminated survival differences between strains and sexes. Septic PAD4-/- mice had reduced IL-10 in the early phase of sepsis, increased lung myeloperoxidase, and exacerbated lung injury compared with septic wild-type mice. CONCLUSION PAD4 deficiency in female mice worsened survival in the fecal-induced peritonitis sepsis model. In both strains, male mice exhibited worse survival compared with their female counterparts. PAD4 deficiency is associated with reduced IL-10, increased neutrophil infiltration, and exacerbated lung injury. Antibiotics eliminated survival differences between strains and sexes.
Collapse
Affiliation(s)
- Erblin Cani
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Dhruva J Dwivedi
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Sean Carlin
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Neha Sharma
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Alex Chen
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Patricia C Liaw
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
13
|
Reis LR, Nascimento RO, Massafera MP, Di Mascio P, Ronsein GE. Investigating neutrophil responses to stimuli: Comparative analysis of reactive species-dependent and independent mechanisms. Redox Biol 2025; 81:103540. [PMID: 40037225 PMCID: PMC11923813 DOI: 10.1016/j.redox.2025.103540] [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: 12/20/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
Neutrophils play a critical role in immune response, using mechanisms as degranulation, phagocytosis, and the release of extracellular DNA together with microbicidal proteins, the so-called neutrophil extracellular traps (NETs), to combat pathogens. Multiple mechanisms might be involved in neutrophil's response to stimuli, but the biochemical characterization of each different pathway is still lacking. In this study, we used superoxide measurements, live-imaging microscopy and high-resolution proteomics to provide a thorough biochemical characterization of the neutrophil's response following activation by two well-known stimuli, namely phorbol-12-myristate-13-acetate (PMA), and ionomycin, a calcium ionophore. Our results demonstrated that although both stimuli induce extracellular DNA release, signals and mediators released by activated cells before this final event were distinct. Thus, PMA-treated neutrophils induce superoxide production, and degranulation of proteins from all granules, especially those derived from secretory vesicles and tertiary granules. On the other hand, ionomycin-treated neutrophils do not stimulate superoxide generation, but induce extensive protein citrullination (also known as arginine deimination), particularly modifying proteins related to actin cytoskeleton organization, nucleus stability, and the NADPH oxidase complex. Interestingly, many of the citrullinated proteins detected in this work were also found to act as autoantigens in autoimmune diseases such as rheumatoid arthritis. These striking differences show neutrophils' response to PMA and ionomycin are two distinct biochemical processes that point towards neutrophils diversification and plasticity responding to the environment. It also provides implications for understanding neutrophil-driven microbial response and potential roles in autoimmune diseases.
Collapse
Affiliation(s)
- Lorenna Rocha Reis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | | | - Mariana Pereira Massafera
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Paolo Di Mascio
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Graziella Eliza Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil.
| |
Collapse
|
14
|
Shin Y, Bae H, Lee C, Rhee I. The dynamic roles of macrophages extracellular traps (METs) in immune regulation. Arch Pharm Res 2025; 48:293-304. [PMID: 40186802 DOI: 10.1007/s12272-025-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Macrophages are crucial to innate immunity, eliminating pathogens and damaged tissues through phagocytosis and modulating immune responses. Recently, macrophage extracellular traps (METs) have been identified as chromatin-based structures composed of DNA and various immune-related proteins. While METs play a defensive role in trapping and neutralizing pathogens, they are also implicated in disease pathology, contributing to chronic inflammation, tissue damage, and immune dysregulation. The precise mechanisms regulating MET formation are still under investigation, but emerging evidence indicates the involvement of various regulatory factors. Dysregulated MET activity has been associated with various diseases, including autoimmune disorders, cancer, and neurological conditions. A deeper understanding of MET mechanisms and their pathological impact may offer novel therapeutic strategies. Given the limited number of reviews and articles on METs, this review provides valuable insights into MET formation, regulatory pathways, and their role in disease progression.
Collapse
Affiliation(s)
- Yunjin Shin
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Hanyoung Bae
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Chaelin Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Inmoo Rhee
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea.
| |
Collapse
|
15
|
Mishra N, Mohs M, Wittmann N, Gross S, Thompson PR, Bossaller L. PLC and PAD2 Regulate Extracellular Calcium-Triggered Release of Macrophage Extracellular DNA Traps. Eur J Immunol 2025; 55:e202350942. [PMID: 40170382 PMCID: PMC11962252 DOI: 10.1002/eji.202350942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/24/2024] [Accepted: 01/20/2025] [Indexed: 04/03/2025]
Abstract
Macrophages can respond to infection or cellular stress by forming inflammasomes or by releasing extracellular traps (ETs) of DNA through METosis. While ETs have been extensively studied in neutrophils, there are fewer studies on METosis. We show that extracellular calcium and LPS enable human monocyte-derived macrophages (hMDM) to release extracellular DNA decorated with myeloperoxidase (MPO) and citrullinated histone, alongside ASC aggregation and IL-1ß maturation, indicating NLRP3 inflammasome activation. Compared with m-CSF differentiated macrophages only gm-CSF differentiated macrophages expressed macrophage elastase (MMP12) and METs released by the latter had significantly more bactericidal activity toward E. coli. Mechanistically, phospholipase C and peptidyl arginine deiminase-2 inhibition attenuate MET release. Interestingly, NLRP3 inflammasome blockade by MCC950 had a significant effect on MET release. Finally, MET release was completely blocked by plasma membrane stabilization by punicalagin. Altogether, we demonstrate that extracellular calcium-activated hMDM extrude DNA, containing citrullinated histones, MPO, MMP12, and ASC specks and released METs kill bacteria independent of hMDM phagocytotic activity. We believe that calcium-activated hMDM adds a physiologically relevant condition to calcium ionophore induced cell death that may be important in autoimmunity.
Collapse
Affiliation(s)
- Neha Mishra
- Section of Rheumatology, Department of Medicine AUniversity Medicine GreifswaldGreifswaldGermany
| | - Magdalena Mohs
- Section of Rheumatology, Department of Medicine AUniversity Medicine GreifswaldGreifswaldGermany
| | - Nico Wittmann
- Section of Pediatric Rheumatology, Department of Pediatric and Adolescent MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Stefan Gross
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
| | - Paul R. Thompson
- Department of Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Medical SchoolWorcesterUSA
| | - Lukas Bossaller
- Section of Rheumatology, Department of Medicine AUniversity Medicine GreifswaldGreifswaldGermany
| |
Collapse
|
16
|
Gao P, Zhou J, Sun L, Liu D. Neutrophil Extracellular Traps in Oral Diseases. Oral Dis 2025; 31:1084-1091. [PMID: 39530338 DOI: 10.1111/odi.15197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE To summarize the current knowledge of the neutrophil extracellular traps (NETs) and its critical role in various oral diseases. METHODS We reviewed the recent research on NETs through PubMed and Web of Science. An analysis of recent research results was summarized from three aspects: NETs induction and formation, functions of NETs, and NETs in oral diseases. RESULTS The relationship between neutrophils and NETs is critical to the body's defense against microbial invasion. NETs can effectively combat pathogens with an anti-inflammatory effect and meanwhile it can contribute to inflammation. Moreover, it can synergize with other immune cells to respond to stimuli, such as pathogens, host-derived mediators, and drugs. It was revealed that NETs play different roles to influence various oral diseases like periodontitis, endodontic infection, oral mucosal diseases, maxillofacial tumors, and many other oral diseases. CONCLUSION The balance between the protective and potentially harmful effects of NETs is a key factor in determining the outcome of infections and inflammatory responses. The role of NETs in oral diseases needs to be further studied to enable better understanding of its role in the different oral diseases.
Collapse
Affiliation(s)
- Pengfei Gao
- Department of Periodontology, Suzhou Stomatological Hospital, Suzhou, Jiangsu, China
| | - Jun Zhou
- Department of Conservative Dentistry, Division of Biomaterials and Engineering, Showa University School of Dentistry, Tokyo, Japan
| | - Lu Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Dayong Liu
- Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Matozzo V, Brunelli N, Cima F. The underrated immune role of bivalve 'serous cells': New insight from inflammatory responses of the Manila clam Ruditapes philippinarum. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110188. [PMID: 39938623 DOI: 10.1016/j.fsi.2025.110188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
This study investigated the immune role of serous cells from the bivalve Ruditapes philippinarum. Histochemical and immunohistochemical assays revealed that the serous cells contained large cytoplasmic vacuoles rich in heparinoid molecules. Heparin and histamine were detected within vacuoles, with distinct spatial distributions, and histoenzymatic assays for serine proteases revealed both tryptase and chymase activity. These findings, together with membrane immunolabelling for c-kit, suggest similarities with vertebrate mast cells. After in vivo bacterial inoculation, serous cells first accumulated at the injury site within 12 h, and 15 h after in vitro treatment, a significant increase in the percentage of serous cells was observed in bacteria-treated samples, supporting targeted responses of proliferation and differentiation following bacterial challenge. Serous cells also underwent marked degranulation following bacterial stimulation, and aggregates of granulocytes, hyalinocytes and serous cells appeared within 1 h of treatment. Extracellular trap formation (ETosis), which is rich in degranulated serous cells and trapped dying bacteria, was observed after 15 h. Serous cells showed nuclear envelope loss and chromatin fragmentation. The extracellular nets were primarily composed of fragmented chromatin and amyloid fibrils forming a scaffold of woven fibres, to which the adhesion of heparin, histamine, and serine proteases occurred after their release into the extracellular environment. To our knowledge, this is the first study that highlights the important role of serous cells in the immune response of bivalves and provides new perspectives for future investigations into the modulation of the inflammatory process.
Collapse
Affiliation(s)
| | | | - Francesca Cima
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
18
|
Varjú I, Tanka-Salamon A, Kolev K. Neutrophil Extracellular Traps: At the Interface of Thrombosis and Comorbidities. Semin Thromb Hemost 2025. [PMID: 40020757 DOI: 10.1055/a-2548-0805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Since their discovery in 2004, neutrophil extracellular traps (NETs) have been at the center of multidisciplinary attention. Although a key tool in neutrophil-mediated immunity, these filamentous, enzyme-enriched DNA-histone complexes can be detrimental to tissues and have been identified as an underlying factor in a range of pathological conditions. Building on more than 20 years of research into NETs, this review places thrombosis, the pathological formation of blood clots, in the spotlight. From this point of view, we discuss the structure and formation of NETs, as well as the interaction of their components with the hemostatic system, dissecting the pathways through which NETs exert their marked effect on formation and the dissolution of thrombi. We pay distinct attention to the latest developments in the research of a key player in NET formation, peptidyl-arginine-deiminase (PAD) enzymes: their types, sources, and potential cross-play with the hemostatic machinery. Besides these molecular details, we elaborate on the link between pathological thrombosis, NETs, and widespread conditions that represent a debilitating public health burden worldwide, such as sepsis and neoplasms. Finally, future implications on the treatment of thrombosis-related conditions will be discussed.
Collapse
Affiliation(s)
- Imre Varjú
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Anna Tanka-Salamon
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Fang J, Ding H, Huang J, Liu W, Hong T, Yang J, Wu Z, Li Z, Zhang S, Liu P, Fang Y, Wu J, Li X, Lin J. Mac-1 blockade impedes adhesion-dependent neutrophil extracellular trap formation and ameliorates lung injury in LPS-induced sepsis. Front Immunol 2025; 16:1548913. [PMID: 40226627 PMCID: PMC11985419 DOI: 10.3389/fimmu.2025.1548913] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Background Sepsis is a common critical condition that can lead to multiple organ injury. Sepsis-induced acute respiratory distress syndrome (ARDS) is frequently an important cause of poor prognosis and is associated with high mortality rates, despite existing therapeutic interventions. Neutrophil infiltration and extracellular traps (NET) are implicated in acute lung injury (ALI) and ARDS following sepsis. As circulating neutrophils infiltrate infected tissues, they come into direct contact with vascular endothelial cells (ECs). Although the ability of NETs to induce endothelial damage is well established, the specific role of direct EC-neutrophil interactions in NET formation and lung injury during sepsis is not fully understood. Methods In this study, NET formation was assessed when neutrophils were co-culture with ECs or separated from them and stimulated with phorbol 12-myristate 13-acetate (PMA), lipopolysaccharide (LPS), lipoteichoic acid (LTA), or septic plasma. Results We found that adhesion of neutrophils on ECs is critical in NET formation in response to LPS, LTA, or septic plasma in vitro. Blocking the macrophage-1 antigen (Mac-1) impeded NET formation, while inhibiting P-selectin glycoprotein ligand-1 (PSGL-1) or leukocyte function-associated antigen-1 (LFA-1) did not. This adhesion-dependent NET formation was reliant on the influx of extracellular calcium and peptidylarginine deiminase 4 (PAD4)-mediated citrullination of histone H3. However, Mac-1 blockade did not alter calcium influx. In a murine model of LPS-induced sepsis, Mac-1 blockade reduced NET release, lowered inflammatory cytokine levels, mitigated endothelial damage, and attenuated lung injury. Conclusion Our findings offer insights into the critical role of EC-neutrophil direct contact in NET formation during sepsis and propose Mac-1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jinhua Fang
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaqi Huang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Wang Liu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Tiantian Hong
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Junxian Yang
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhiwei Wu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhuo Li
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shiying Zhang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Peimin Liu
- Department of Nephrology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Fang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jianhua Wu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiangguo Lin
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
20
|
Jiang X, Li Q, Huang R, Qian Y, Jiang Y, Liu T, Wang Y, Hu K, Huang J, Huang W, Liu Q, Wei Z, Zhang H, Yu X. Giardia duodenalis triggered neutrophil extracellular traps in goats. Immunobiology 2025; 230:152894. [PMID: 40185010 DOI: 10.1016/j.imbio.2025.152894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
Giardia duodenalis is a globally distributed zoonotic parasite primarily transmitted through the fecal-oral route, infecting various vertebrates, and the infection of which is prevalent in goats. Immune cells play a crucial role in pathogens invasion, and neutrophil extracellular traps (NETs) released by neutrophils serve as a non-specific defense mechanism against pathogens including parasites. In this study, we investigated the characteristics, components, and molecular mechanisms of goat NETs upon stimulation with G. duodenalis trophozoites. This study demonstrates that G. duodenalis trigger dose-dependent NETs formation in goat neutrophils, composed of DNA, citrullinated histone H3 (CitH3), and neutrophil elastase (NE). Reactive oxygen species (ROS) accumulation synchronizes with NETosis during G. duodenalis infection. Inhibitor experiments confirmed that G. duodenalis-induced NETs and ROS production depend on TLR2/4 signaling and require NADPH oxidase (NOX), ERK1/2, and p38 MAPK activation. This work identifies TLR2/4, NOX, ERK1/2, and p38 MAPK pathways as key regulators of NETs/ROS coordination during G. duodenalis infection, providing the first evidence of G. duodenalis-triggered NETs in goats. The findings highlight NETs as critical components of anti-G. duodenalis immunity and suggest potential for NETs-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Xi Jiang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Qiaoyu Li
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Rongsheng Huang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yuxiao Qian
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yuqian Jiang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Tingting Liu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yiwen Wang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Kairao Hu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Jing Huang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Wenlong Huang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Quan Liu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhengkai Wei
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Haoji Zhang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Xingang Yu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China.
| |
Collapse
|
21
|
Saffarian M, Romano JD, Grigg ME, Coppens I. A major Toxoplasma serine protease inhibitor protects the parasite against gut-derived serine proteases and NETosis damage. J Biol Chem 2025; 301:108457. [PMID: 40154616 DOI: 10.1016/j.jbc.2025.108457] [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: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
Toxoplasmosis is a life-threatening opportunistic infection in immunocompromised patients, caused by the parasite Toxoplasma gondii. Infection is initiated through oral ingestion of Toxoplasma cysts that must survive the harsh environment of the gut to undergo excystation. Released parasites invade intestinal epithelial cells and then disseminate throughout tissues for encystation, mainly in the brain. How Toxoplasma escapes destruction mediated by gastrointestinal proteases is poorly understood. T. gondii has nine genes encoding serine protease inhibitor proteins (TgPIs). TgPI-1 is highly expressed across all Toxoplasma strains and developmental stages and contains three domains for binding to various serine proteases. Here, we explore the role of TgPI-1 in protecting Toxoplasma against serine proteases in the gut and neutrophil-derived proteases in the lamina propria. TgPI-1 localizes to the parasite plasma membrane and cyst wall. We generated ΔTgPI-1 parasites, and the mutant is more sensitive to neutrophil elastase (NE), trypsin and chymotrypsin than WT. Neutrophils exposed to Toxoplasma release neutrophil extracellular traps (NET) with strain-dependent morphologies, ranging from spiky to extended cloudy. TgPI-1 was detected on NET containing NE, and ΔTgPI-1 parasites are more susceptible to destruction by NETosis. In mice, ΔTgPI-1 parasites exhibit reduced infectivity, poor dissemination to abdominal organs, and lower cyst burden in the brain. These findings shed light on a strategy employed by Toxoplasma to counteract enzymatic antimicrobial defenses in gut tissues, highlighting potential avenues for controlling tissue dissemination of this medically significant parasite.
Collapse
Affiliation(s)
- Maryam Saffarian
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Julia D Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA.
| |
Collapse
|
22
|
Yuan Y, Sun C, Liu X, Hu L, Wang Z, Li X, Zhang J, Li D, Zhang X, Wu M, Liu L. The Role of Neutrophil Extracellular Traps in Atherosclerosis: From the Molecular to the Clinical Level. J Inflamm Res 2025; 18:4421-4433. [PMID: 40162077 PMCID: PMC11955173 DOI: 10.2147/jir.s507330] [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: 11/27/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory condition that is typified by the deposition of lipids and the subsequent inflammation of medium and large arteries. Neutrophil extracellular traps (NETs) are fibrous meshworks of DNA, histones, and granzymes expelled by activated neutrophils in response to a variety of pathogenic conditions. In addition to their role in pathogen eradication, NETs have been demonstrated to play a pivotal role in the development of atherosclerosis. This article presents a review of the bidirectional interactions in which atherosclerosis-related risk factors stimulate the formation of NETs, which in turn support disease progression. This article emphasizes the involvement of NETs in the various stages of atherogenesis and development, influencing multiple factors such as the vascular endothelium, platelets, the inflammatory milieu, and lipid metabolism. The findings of this study offer new insights and avenues for further investigation into the processes underlying the formation and regulation of the vascular inflammatory microenvironment in atherosclerosis. Finally, potential targeted therapeutic strategies for NETs are discussed to facilitate their progression to clinical practice (Graphical Abstract).
Collapse
Affiliation(s)
- Yongfang Yuan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Changxin Sun
- Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Lanqing Hu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Zeping Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Xiaoya Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Jingyi Zhang
- Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Dexiu Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Xiaonan Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Longtao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, People’s Republic of China
| |
Collapse
|
23
|
HAO X, FENG Y, LU A, SUN Y, XIA J, MEI X, FENG L, JIANG M, WANG B, YANG H. [Research Progress of Neutrophil Extracellular Traps in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:201-212. [PMID: 40210480 PMCID: PMC11986667 DOI: 10.3779/j.issn.1009-3419.2025.106.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Indexed: 04/12/2025]
Abstract
Neutrophil extracellular traps (NETs), intricate reticular structures released by activated neutrophils, play a pivotal regulatory role in the pathogenesis of malignant tumors. Lung cancer is one of the most prevalent malignancies globally, with persistently high incidence and mortality rates. Recent studies have revealed that NETs dynamically modulate the tumor microenvironment through unique pathological mechanisms, exhibiting complex immunoregulatory characteristics during the progression of lung cancer, and this discovery has increasingly become a focal point in tumor immunology research. This paper provides a comprehensive review of the latest advancements in NETs research related to lung cancer, offering an in-depth analysis of their impact on lung cancer progression, their potential diagnostic value, and the current state of research on targeting NETs for lung cancer prevention and treatment. The aim is to propose novel strategies to enhance therapeutic outcomes and improve the prognosis for lung cancer patients.
.
Collapse
|
24
|
Nakazawa D, Masuda S, Nishibata Y, Watanabe-Kusunoki K, Tomaru U, Ishizu A. Neutrophils and NETs in kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00944-3. [PMID: 40102634 DOI: 10.1038/s41581-025-00944-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Neutrophils, conventionally regarded as a homogeneous immune cell population, have emerged as a heterogeneous group of cells with distinct gene profiles and immune properties. Activated neutrophils release a spectrum of bioactive substances, including cytokines, chemokines, proteolytic enzymes, reactive oxygen species and neutrophil extracellular traps (NETs), which are composed of decondensed DNA and antimicrobial proteins. NETs have a pivotal role in innate immunity, including in preventing the ascent of uropathogenic bacteria into the kidneys, as they efficiently trap pathogenic microorganisms. However, although indispensable for defence against pathogens, NETs also pose risks of self-damage owing to their cytotoxicity, thrombogenicity and autoantigenicity. Accordingly, neutrophils and NETs have been implicated in the pathogenesis of various disorders that affect the kidneys, including acute kidney injury, vasculitis, systemic lupus erythematosus, thrombotic microangiopathy and in various aetiologies of chronic kidney disease. Pathological alterations in the glomerular vascular wall can promote the infiltration of neutrophils, which can cause tissue damage and inflammation through their interactions with kidney-resident cells, including mesangial cells and podocytes, leading to local cell death. Targeting neutrophil activation and NET formation might therefore represent a new therapeutic strategy for these conditions.
Collapse
Affiliation(s)
- Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yuka Nishibata
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Kanako Watanabe-Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
25
|
Skjølberg C, Degani L, Sileikaite-Morvaközi I, Hawkins CL. Oxidative modification of extracellular histones by hypochlorous acid modulates their ability to induce β-cell dysfunction. Free Radic Biol Med 2025; 230:209-221. [PMID: 39956473 DOI: 10.1016/j.freeradbiomed.2025.02.018] [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: 01/09/2025] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 02/18/2025]
Abstract
Histones are nuclear proteins that play a key role in chromatin assembly and regulation of gene expression by their ability to bind to DNA. Histones can also be released from cells owing to necrosis or extracellular trap release from neutrophils (NETs) and other immune cells. The presence of histones in the extracellular environment has implications for many pathologies, including diabetes mellitus, owing to the cytotoxic nature of these proteins, and their ability to promote inflammation. NETs also contain myeloperoxidase, a defensive enzyme that produces hypochlorous acid (HOCl), to kill pathogens, but also readily damages host proteins. In this study, we examined the reactivity of histones with and without HOCl modification, with a pancreatic β-cell model. Exposure of β-cells to histones resulted in a loss of metabolic activity and cell death by a combination of apoptosis and necrosis. This toxicity was increased on pretreatment of the β-cells with tumour necrosis factor α and interleukin 1β. Histones upregulated endoplasmic reticulum (ER) stress genes, including the pro-apoptotic transcription factor CHOP. There was also evidence for alterations to the cellular redox environment and upregulation of antioxidant gene expression. However, downregulation of insulin-associated genes and insulin was observed. Interestingly, modification of the histones with HOCl reduced their toxicity and altered the patterns of gene expression observed, and a further decrease in the expression of insulin-associated genes was observed. These findings could be relevant to the development of Type 2 diabetes, where low-grade inflammation favours NET release, resulting in elevated histones in the circulation.
Collapse
Affiliation(s)
- Clara Skjølberg
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Laura Degani
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Inga Sileikaite-Morvaközi
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
26
|
Li S, Dong P, Wu X, Kang Z, Yan G. Global trends in tumor-associated neutrophil research: a bibliometric and visual analysis. Front Immunol 2025; 16:1478092. [PMID: 40160822 PMCID: PMC11949894 DOI: 10.3389/fimmu.2025.1478092] [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: 08/09/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Background Tumor-associated neutrophils (TANs) play crucial roles in tumor progression, immune response modulation, and the therapeutic outcomes. Despite significant advancements in TAN research, a comprehensive bibliometric analysis that objectively presents the current status and trends in this field is lacking. This study aims to fill this gap by visually analyzing global trends in TANs research using bibliometric and knowledge mapping techniques. Methods We retrieved articles and reviews related to TANs from the Web of Science core collection database, spanning the period from 2012 to2024. The data was analyzed using bibliometric tools such as Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio) to identify key trends, influential countries and institutions, collaborative networks. and citation patterns. Results A total of 6l5 publications were included in the bibliometric analysis, showing a significant upward trend in TANs research over the last two decades. The United States and China emerged as the leading contributors with the highest number of publications and citations. The journal with the most publications in this field is Frontiers in Immunology, Prominent authors such as Fridlender ZG was identified as the key contributor, with his works frequently cited. The analysis highlighted major research themes. including the role of TANs in tumor microenvironment modulation, their dual functions in tumor promotion and suppression, and the exploration of TANs-targeted therapies, Emerging research hotspots include studies on TANs plasticity and their interactions with other immune cells. Conclusion This study is the first to employ bibliometric methods to visualize trends and frontiers in TANs research. The findings provide valuable insights into the evolution of the field, highlighting critical areas for future investigation and potential collaborative opportunities. This comprehensive analysis serves as a crucial resource for researchers and practitioners aiming to advance TAN research and its application in cancer therapy.
Collapse
Affiliation(s)
- Shaodong Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Peng Dong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhenhua Kang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Guoqiang Yan
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Hellenthal KEM, Thomas K, Ludwig N, Cappenberg A, Schemmelmann L, Tekath T, Margraf A, Mersmann S, Henke K, Rossaint J, Zarbock A, Amini W. Glutamine modulates neutrophil recruitment and effector functions during sterile inflammation. J Leukoc Biol 2025; 117:qiae243. [PMID: 39504570 DOI: 10.1093/jleuko/qiae243] [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: 07/18/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024] Open
Abstract
During sterile inflammation, tissue damage induces excessive activation and infiltration of neutrophils into tissues, where they critically contribute to organ dysfunction. Tight regulation of neutrophil migration and their effector functions is crucial to prevent overshooting immune responses. Neutrophils utilize more glutamine, the most abundant free α-amino acid in the human blood, than other leukocytes. However, under inflammatory conditions, the body's requirements exceed its ability to produce sufficient amounts of glutamine. This study investigates the impact of glutamine on neutrophil recruitment and their key effector functions. Glutamine treatment effectively reduced neutrophil activation by modulating β2-integrin activity and chemotaxis in vitro. In a murine in vivo model of sterile inflammation induced by renal ischemia-reperfusion injury, glutamine administration significantly attenuated neutrophil recruitment into injured kidneys. Transcriptomic analysis revealed, glutamine induces transcriptomic reprograming in murine neutrophils, thus improving mitochondrial functionality and glutathione metabolism. Further, glutamine influenced key neutrophil effector functions, leading to decreased production of reactive oxygen species and formation of neutrophil extracellular traps. Mechanistically, we used a transglutaminase 2 inhibitor to identify transglutaminase 2 as a downstream mediator of glutamine effects on neutrophils. In conclusion, our findings suggest that glutamine diminishes activation and recruitment of neutrophils and thus identify glutamine as a potent means to curb overshooting neutrophil responses during sterile inflammation.
Collapse
Affiliation(s)
- Katharina E M Hellenthal
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Katharina Thomas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Nadine Ludwig
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
- Department of Cardiothoracic Surgery, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Anika Cappenberg
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Lena Schemmelmann
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Tobias Tekath
- Institute of Medical Informatics, University of Muenster, Albert-Schweitzer-Campus 1, Building A11, 48149 Muenster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Sina Mersmann
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Katharina Henke
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Wida Amini
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| |
Collapse
|
28
|
Yang L, Guo R, Liu H, Chen B, Li C, Liu R, Liao S, Xie Q, Yin G. Mechanism of antiphospholipid antibody-mediated thrombosis in antiphospholipid syndrome. Front Immunol 2025; 16:1527554. [PMID: 40181965 PMCID: PMC11966034 DOI: 10.3389/fimmu.2025.1527554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by the occurrence of thrombotic or obstetrical events in patients with persistent antiphospholipid antibodies (aPL). Thrombotic events, the primary pathological hallmarks and clinical manifestations, are among the leading causes of mortality in APS. Our understanding of the mechanism underlying APS-related thrombosis has significantly advanced in recent years. The presence of aPL, particularly anti-β2-glycoprotein I (anti-β2GPI) antibodies, is a major driver of thrombosis. The proposed pathophysiological mechanisms of aPL-mediated pro-thrombotic events can be broadly categorized into three types: disruption of anticoagulant reactions and fibrinolysis, interference with coagulation cascade cells, and complement activation. A triggering 'second hit' is typically necessary to initiate thrombosis. The development of animal models of APS has further refined our understanding of the role of aPL in thrombosis. In this review, we focused on the role of β2GPI-dependent aPL in thrombosis of thrombotic APS.
Collapse
Affiliation(s)
- Leiyi Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruibing Guo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Changpei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiting Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyi Liao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Wise AD, TenBarge EG, Mendonça JDC, Mennen EC, McDaniel SR, Reber CP, Holder BE, Bunch ML, Belevska E, Marshall MG, Vaccaro NM, Blakely CR, Wellawa DH, Ferris J, Sheldon JR, Bieber JD, Johnson JG, Burcham LR, Monteith AJ. Mitochondria sense bacterial lactate and drive release of neutrophil extracellular traps. Cell Host Microbe 2025; 33:341-357.e9. [PMID: 40020664 PMCID: PMC11955204 DOI: 10.1016/j.chom.2025.02.003] [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: 06/11/2024] [Revised: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Neutrophils induce oxidative stress, creating a harsh phagosomal environment. However, Staphylococcus aureus can survive these conditions, requiring neutrophils to deploy mechanisms that sense bacterial persistence. We find that staphylococcal lactate is a metabolic danger signal that triggers neutrophil extracellular trap release (NETosis). Neutrophils coordinate mitochondria in proximity to S. aureus-containing phagosomes, allowing transfer of staphylococcal lactate to mitochondria where it is rapidly converted into pyruvate and causes mitochondrial reactive oxygen species, a precursor to NETosis. Similar results were observed in response to phylogenetically distinct bacteria, implicating lactate accumulation as a broad signal triggering NETosis. Furthermore, patients with systemic lupus erythematosus (SLE) are more susceptible to bacterial infections. We find that SLE neutrophils cannot sense bacterial lactate impairing their capacity to undergo NETosis upon S. aureus infection but initiate aberrant NETosis triggered by apoptotic debris. Thus, neutrophils adapt mitochondria as sensory organelles that detect bacterial metabolic activity and dictate downstream antibacterial processes.
Collapse
Affiliation(s)
- Ashley D Wise
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Eden G TenBarge
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Ellie C Mennen
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Sarah R McDaniel
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Callista P Reber
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Bailey E Holder
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Madison L Bunch
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Eva Belevska
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Nicole M Vaccaro
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | | - Dinesh H Wellawa
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine and Infectious Disease Organization, Saskatoon, SK, Canada
| | - Jennifer Ferris
- Division of Rheumatology, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Jessica R Sheldon
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine and Infectious Disease Organization, Saskatoon, SK, Canada
| | - Jeffry D Bieber
- Division of Rheumatology, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Jeremiah G Johnson
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lindsey R Burcham
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | - Andrew J Monteith
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA; Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
30
|
Xue H, Xie R, Wang Z, Fan W, Wei Y, Zhang L, Zhao D, Song Z. Coordination of Neutrophil and Apoptosis-Inducing Ligand in Inflammatory Diseases. J Inflamm Res 2025; 18:3607-3621. [PMID: 40099000 PMCID: PMC11911651 DOI: 10.2147/jir.s506807] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
As the most abundant innate immune cells, neutrophils play a key role in host's anti-infective activity and tissue damage/repair process of sterile inflammation. Due to the restriction of apoptosis and other regulatory mechanisms, neutrophils have a short survival time in vivo. Because of the death domain of cytoplasmic regions, some members of tumor necrosis factor receptor superfamily (TNFRSF) are defined as death receptors, such as TNFR-I, Fas and DR4/DR5. TNF-α, FasL and TRAIL, which are known as apoptosis-inducing ligand, can bind to death receptors and activate intracellular apoptosis pathways to induce apoptosis. Accumulating studies found that these three apoptosis-inducing ligands play an important role in the immune system by coordinating with neutrophil, which including neutrophil recruitment/infiltration and function performing. In this review, we summarize existing studies targeting neutrophils as diagnosis and treatment for diseases, and focus on the involvement of neutrophils which regulated by apoptosis-inducing ligands in inflammatory diseases under current cognition.
Collapse
Affiliation(s)
- Hanyu Xue
- The First Affiliated Hospital of Henan University, School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Ran Xie
- School of Medical Technology, Shangqiu Medical College, Shangqiu, 476000, People's Republic of China
| | - Zhiwei Wang
- Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475000, People's Republic of China
| | - Wenqian Fan
- School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Yinxiang Wei
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Lijie Zhang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Dan Zhao
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Zhiming Song
- Department of Cardiology, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
- Kaifeng Key Laboratory for Modulation and Rehabilitation of Cardiac Function, The First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| |
Collapse
|
31
|
Li Y, Cao Z, Liu J, Qiang R, Wang J, Lyu W. Current perspectives and trends of neutrophil extracellular traps in organ fibrosis: a bibliometric and visualization study. Front Immunol 2025; 16:1508909. [PMID: 40109341 PMCID: PMC11920176 DOI: 10.3389/fimmu.2025.1508909] [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: 10/10/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
New insights into the role of immune responses in the fibrosis process provide valuable considerations for the treatment of organ fibrotic diseases. Neutrophil extracellular traps (NETs) represent a novel understanding of neutrophil functions, and their involvement in organ fibrotic diseases has garnered widespread attention in recent years. This study aims to conduct a bibliometric analysis and literature review focusing on the mechanisms by which NETs participate in fibrotic diseases. Specifically, we utilized a bibliometric dataset that includes 220 papers published in 139 journals, originating from 425 organizations across 39 countries, with a total citation count of 12,301. Keyword co-occurrence analysis indicates that the research focus on the mechanisms of NETs in organ fibrosis is likely to center on NETosis, immune responses, immune thrombosis, inflammation, and tissue damage associated with NET formation. In conclusion, our findings underscore the current status and emerging trends in NET research related to organ fibrosis, offering novel insights into the mechanisms by which NETs contribute to the pathogenesis of fibrotic diseases, as well as potential therapeutic strategies.
Collapse
Affiliation(s)
- Yanbo Li
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Zhengmin Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Rui Qiang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Shunyi Hospital, Beijing, China
| | - Jiuchong Wang
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenliang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Liao R, Wang L, Zeng J, Tang X, Huang M, Kantawong F, Huang Q, Mei Q, Huang F, Yang Y, Liao B, Wu A, Wu J. Reactive oxygen species: Orchestrating the delicate dance of platelet life and death. Redox Biol 2025; 80:103489. [PMID: 39764976 PMCID: PMC11759559 DOI: 10.1016/j.redox.2025.103489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Platelets, which are vital for blood clotting and immunity, need to maintain a delicately balanced relationship between generation and destruction. Recent studies have highlighted that reactive oxygen species (ROS), which act as second messengers in crucial signaling pathways, are crucial players in this dance. This review explores the intricate connection between ROS and platelets, highlighting their dual nature. Moderate ROS levels act as potent activators, promoting megakaryocyte (MK) differentiation, platelet production, and function. They enhance platelet binding to collagen, increase coagulation, and directly trigger cascades for thrombus formation. However, this intricate role harbors a double-edged sword. Excessive ROS unleash its destructive potential, triggering apoptosis and reducing the lifespan of platelets. High levels can damage stem cells and disrupt vital redox-dependent signaling, whereas uncontrolled activation promotes inappropriate clotting, leading to thrombosis. Maintaining a precise balance of ROS within the hematopoietic microenvironment is paramount for optimal platelet homeostasis. While significant progress has been made, unanswered questions remain concerning specific ROS signaling pathways and their impact on platelet disorders. Addressing these questions holds the key to unlocking the full potential of ROS-based therapies for treating platelet-related diseases such as thrombocytopenia and thrombosis. This review aims to contribute to this ongoing dialog and inspire further exploration of this exciting field, paving the way for novel therapeutic strategies that harness the benefits of ROS while mitigating their dangers.
Collapse
Affiliation(s)
- Rui Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jing Zeng
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoqin Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Miao Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qianqian Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qibing Mei
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Feihong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
33
|
Carvalho AS, Pereira-Silva GC, Andrade JMP, Ferreira WS, Weissmüller G, Saraiva EM, Da Poian AT. DNA Extracellular Traps Released by Mayaro Virus-Infected Macrophages Act as a Platform for Virus Dissemination. J Med Virol 2025; 97:e70262. [PMID: 40007117 DOI: 10.1002/jmv.70262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Mayaro virus (MAYV) is an arthritogenic arbovirus that causes a debilitating illness that can progress to a chronic rheumatic disease characterized by persistent viral replication in macrophages within joint tissues. Here, we report that MAYV-infected macrophages release decondensed DNA traps (DNA extracellular traps, DETs) through a mechanism driven by the production of reactive oxygen species and peptidyl arginine deiminase activation, resembling the classical mechanism of pathogen clearance by activated neutrophils. Unlike traditional pathogen clearance observed for NETs released by neutrophils, MAYV-induced DETs did not inactivate the virus. Instead, DET-ensnared viruses are internalized by neighboring uninfected macrophages, increasing the number of infected cells. Collectively, these findings suggest that MAYV-containing DETs act as a "Trojan horse" that facilitates viral dissemination within inflamed tissues, connecting macrophage-mediated inflammatory response to viral persistence in the articular tissue in chronic MAYV disease.
Collapse
Affiliation(s)
- Ana S Carvalho
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gean Carlo Pereira-Silva
- Institudo de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia M P Andrade
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wellington S Ferreira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto Weissmüller
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elvira M Saraiva
- Institudo de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Pisani F, Porciani C, Croia C, Pucino V, Virdis A, Puxeddu I, Migliorini P, Pratesi F. Novel Methods for the Analysis of Serum NET Remnants: Evaluation in Patients with Severe COVID-19. Int J Mol Sci 2025; 26:2221. [PMID: 40076841 PMCID: PMC11899909 DOI: 10.3390/ijms26052221] [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: 01/17/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophil extracellular traps (NETs) are web-like structures composed of chromatin and proteins from neutrophil granules. Several studies highlight the heterogeneity of NETs, underscoring the challenges associated with their detection. In patients with COVID-19, high levels of NET fragments, called NET remnants, are detected in the circulation but also in alveoli and bronchioles. NET remnants are usually measured as complexes of DNA and myeloperoxidase (DNA-MPO). Taking advantage of proteomic data on NET composition, we developed new solid-phase assays to detect NET remnants, measuring complexes of DNA with alpha enolase (DNA-eno) or calprotectin (DNA-cal). The two assays were compared with the DNA-MPO test for the detection of in vitro-generated NET and serum NET remnants; all of them showed similar sensitivity in the detection of in vitro-generated NET. In an analysis of 40 patients with severe COVID-19 and 25 healthy subjects, the results of the three assays were highly correlated, and all detected significantly higher levels of NET remnants in patient sera. Moreover, the level of NET remnants correlated with impaired gas exchange and increased with the progressive decline of pulmonary function. The proposed assays thus represent a novel tool with which to evaluate NETosis; using antibodies to different NET constituents may allow their fingerprinting in different disorders.
Collapse
Affiliation(s)
- Francesco Pisani
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Caterina Porciani
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Cristina Croia
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Valentina Pucino
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Agostino Virdis
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Ilaria Puxeddu
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Paola Migliorini
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (F.P.); (C.P.); (C.C.); (V.P.); (I.P.); (P.M.)
| | - Federico Pratesi
- Department of Translational Medicine and NTMS, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
35
|
Zawrotniak M, Satala D, Juszczak M, Bras G, Rapala-Kozik M. Candida albicans aspartyl protease (Sap6) inhibits neutrophil function via a "Trojan horse" mechanism. Sci Rep 2025; 15:6946. [PMID: 40011643 PMCID: PMC11865311 DOI: 10.1038/s41598-025-91425-x] [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: 09/04/2024] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
Candida albicans, a prevalent fungal pathogen, employs aspartyl proteases such as Sap6 to evade immune defenses, challenging our understanding of host‒pathogen interactions. This research examined the impact of Sap6 on neutrophil responses, which are crucial for innate immunity. Employing flow cytometry and fluorescence microscopy, we explored how Sap6 affects neutrophil functions, particularly by focusing on reactive oxygen species (ROS) production, neutrophil extracellular traps release (NETosis), and apoptosis. Our findings revealed Sap6's unique ability to bind and internalize in neutrophils, significantly attenuating ROS production through proteolytic damage to NADPH oxidase, resulting in blocking the ROS-dependent NETosis pathway. This disruption in neutrophil functions by Sap6 suggested the presence of a 'Trojan horse' mechanism by C. albicans. This mechanism reveals a sophisticated immune evasion strategy, shedding light on fungal pathogenicity and host immune interactions. Understanding fungal proteases in immune modulation could inspire new therapeutic approaches for fungal infections.
Collapse
Affiliation(s)
- Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Magdalena Juszczak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Grażyna Bras
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
36
|
Li H, Shan W, Zhao X, Sun W. Neutrophils: Linking Inflammation to Thrombosis and Unlocking New Treatment Horizons. Int J Mol Sci 2025; 26:1965. [PMID: 40076593 PMCID: PMC11901051 DOI: 10.3390/ijms26051965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophils play a key role in inflammatory responses and thrombosis, but their complex interactions in disease pathogenesis are not fully understood. This review examines the multifaceted roles of neutrophils, focusing on their activation, cytokine release, and formation of neutrophil extracellular traps (NETs), which contribute to host defense and thrombosis. We discuss the interaction between inflammation and coagulation, the direct effect of neutrophils on thrombus stability, and their involvement in pathological thrombotic diseases. The therapeutic potential of neutrophil drug loading in the treatment of thrombosis, as well as the clinical implications and future research directions, are highlighted. The aim of this review is to gain insight into the critical neutrophil-inflammation-thrombus axis and its potential as a therapeutic target for thrombotic diseases and to suggest possible directions for neutrophil-loaded drug therapy for thrombosis.
Collapse
Affiliation(s)
| | | | | | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.L.); (W.S.); (X.Z.)
| |
Collapse
|
37
|
Sennett C, Pula G. Trapped in the NETs: Multiple Roles of Platelets in the Vascular Complications Associated with Neutrophil Extracellular Traps. Cells 2025; 14:335. [PMID: 40072064 PMCID: PMC11898727 DOI: 10.3390/cells14050335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Neutrophil extracellular traps (NETs) have received significant attention in recent years for their role in both the immune response and the vascular damage associated with inflammation. Platelets have been described as critical components of NETs since the initial description of this physio-pathological response of neutrophils. Platelets have been shown to play a dual role as responders and also as stimulators of NETs. The direct interaction with DNA leads to the entrapment of platelets into NETs, a phenomenon that significantly contributes to the thrombotic complications of inflammation and neutrophil activation, while the direct and paracrine stimulation of neutrophils by platelets has been shown to initiate the process of NET formation. In this review, we provide a comprehensive description of our current understanding of the molecular mechanisms underlying the entrapping of platelets into NETs and, in parallel, the platelet-driven cellular responses promoting NET formation. We then illustrate established examples of the contribution of NETs to vascular pathologies, describe the important questions that remain to be answered regarding the contribution of platelets to NET formation and NET-dependent cardiovascular complication, and highlight the fundamental steps taken towards the application of our understanding of platelets' contribution to NETs for the development of novel cardiovascular therapies.
Collapse
Affiliation(s)
| | - Giordano Pula
- Biomedical Institute for Multimorbidity (BIM), Hull York Medical School (HYMS), University of Hull, Hull HU6 7RX, UK
| |
Collapse
|
38
|
Zhang Y, Ye J, Sun S, Li R, Tang S, Wang M, Sun G. Role of platelets and NETs in arterial thrombosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03921-6. [PMID: 39992420 DOI: 10.1007/s00210-025-03921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Arterial thrombosis is one of the main causes of mortality and mortality worldwide. Platelets are effectively targeted by antithrombotic strategies. However, current antiplatelet agents are often associated with a bleeding risk and single antiplatelet agent may not completely prevent thrombosis. Platelets, neutrophils, and neutrophil extracellular traps (NETs) have been found to play crucial synergistic roles in the pathological process of arterial thrombosis in recent years. Platelets play a key regulatory role in the formation of NETs, and NETs can enhance platelet aggregation and activation, further aggravating the process of arterial thrombosis. Targeting the interaction mechanisms of platelets and NETs may provide a promising approach to better prevent arterial thrombosis. This review highlights the current insight in the interaction of platelets and neutrophil-forming NETs and their mechanisms involved in the process of arterial thrombosis. Finally, we discuss the potential of interventions targeting platelets and NETs to treat arterial thrombosis.
Collapse
Affiliation(s)
- Yaqi Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shiyi Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ruoyun Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shuang Tang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
39
|
Zhou X, Jin Y, Zhu Y, Luo X, Li S, Shen W, Wu R. The Role of Crosstalk between Nets and Keratinocytes in Skin Immunity. J Invest Dermatol 2025:S0022-202X(25)00012-0. [PMID: 39985552 DOI: 10.1016/j.jid.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 02/24/2025]
Abstract
The skin is the principal barrier against pathogens. Skin-resident cells, especially keratinocytes, play essential roles in skin immunity. Damage to the integrity of the skin barrier triggers the localized release of proinflammatory factors from keratinocytes, which attract neutrophils. These infiltrating neutrophils in turn release cytokines to modulate keratinocyte function, thereby amplifying skin inflammation. In addition, neutrophils produce neutrophil extracellular traps in a process called NETosis. Notably, crosstalk between neutrophils and keratinocytes is a prominent feature of skin infection eradication and autoimmune disorder development. In this paper, we review research progress on neutrophil extracellular traps in cutaneous immunity, with a particular emphasis on their modulation of keratinocytes. Moreover, we discuss the implications of neutrophil heterogeneity for immune defense and disease development and treatment.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Yi Jin
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Yanshan Zhu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Xin Luo
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Siying Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Weiyun Shen
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Ruifang Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China.
| |
Collapse
|
40
|
Alekseeva LA, Sen’kova AV, Sounbuli K, Savin IA, Zenkova MA, Mironova NL. Pulmozyme Ameliorates LPS-Induced Lung Fibrosis but Provokes Residual Inflammation by Modulating Cell-Free DNA Composition and Controlling Neutrophil Phenotype. Biomolecules 2025; 15:298. [PMID: 40001601 PMCID: PMC11853346 DOI: 10.3390/biom15020298] [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: 11/26/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Pulmonary fibrosis, a chronic progressive lung disorder, can be the result of previous acute inflammation-associated lung injury and involves a wide variety of inflammatory cells, causing the deposition of extracellular matrix (ECM) components in the lungs. Such lung injury is often associated with excessive neutrophil function and the formation of DNA networks in the lungs, which are also some of the most important factors for fibrosis development. Acute lung injury with subsequent fibrosis was initiated in C57Bl/6 mice by a single intranasal (i.n.) administration of LPS. Starting from day 14, human recombinant DNase I in the form of Pulmozyme for topical administration was instilled i.n. twice a week at a dose of 50 U/mouse. Cell-free DNA (cfDNA), DNase activity, and cell content were analyzed in blood serum and bronchoalveolar lavage fluid (BALF). Inflammatory and fibrotic changes in lung tissue were evaluated by histological analysis. The gene expression profile in spleen-derived neutrophils was analyzed by RT-qPCR. We demonstrated that Pulmozyme significantly reduced connective tissue expansion in the lungs. However, despite the reliable antifibrotic effect, complete resolution of inflammation in the respiratory system of mice treated with Pulmozyme was not achieved, possibly due to enhanced granulocyte recruitment and changes in the nuclear/mitochondrial cfDNA balance in the BALF. Moreover, Pulmozyme introduction caused the enrichment of the spleen-derived neutrophil population by those with an unusual phenotype, combining pro-inflammatory and anti-inflammatory features, which can also maintain lung inflammation. Pulmozyme can be considered a promising drug for lung fibrosis management; however, the therapy may be accompanied by residual inflammation.
Collapse
Affiliation(s)
- Ludmila A. Alekseeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Khetam Sounbuli
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova St., 1, Novosibirsk 630090, Russia
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| | - Nadezhda L. Mironova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), Lavrentiev Ave., 8, Novosibirsk 630090, Russia; (L.A.A.); (A.V.S.); (K.S.); (I.A.S.); (M.A.Z.)
| |
Collapse
|
41
|
Porfírio
do Nascimento PR, Mendes-Aguiar CO, Morais IC, Rodrigues Neto JF, Wilson ME, Jerônimo SMB. Neutrophil- Leishmania infantum Interaction Induces Neutrophil Extracellular Traps, DAMPs, and Inflammatory Molecule Release. ACS Infect Dis 2025; 11:483-492. [PMID: 39887021 PMCID: PMC11833875 DOI: 10.1021/acsinfecdis.4c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Neutrophils, the first cells to arrive at infection sites, release neutrophil extracellular traps (NETs) comprising nuclear and/or mitochondrial DNA webs decorated with proteins. Similar to other parasites, Leishmania infantum induces NET extrusion. However, our understanding of NET formation and neutrophil fate after NET release in a Leishmania infection context is limited. Our study aimed to determine the DNA origin of the NET scaffolds released by human neutrophils in response to chemical or L. infantum stimulation. Neutrophils were incubated with PMA, PHA, LPS, or L. infantum, followed by DNA and elastase activity quantification; additionally, we evaluated the source of DNA that composes NETs. Neutrophil viability was evaluated by annexin-V/7AAd labeling. Expression of IL6, TNFA, IL10, CXCL1, CXCL8, and FPR1 in response to the L. infantum interaction was assessed. Neutrophils incubated with chemicals or L. infantum released NETs. However, neutrophils stimulated by the chemicals showed lower viability after 1 h in comparison to neutrophils incubated with parasites. NETs from chemically stimulated neutrophils were mainly composed of nuclear DNA. Conversely, the NET induced by the parasites was of mitochondrial DNA origin and had no leishmanicidal activity. After 4 h of parasite stimulation, neutrophils peak the expression of genes such as IL6, TNFA, CXCL1, CXCL8, and FPR1. Our study demonstrates that neutrophils produce NETs after chemical or L. infantum exposure. Although they are not toxic to the parasite, NETs are released as danger signals. These findings support the role of neutrophils in releasing signaling molecules, which influence the inflammatory environment in which the infectious process occurs.
Collapse
Affiliation(s)
| | | | - Ingryd Câmara Morais
- Institute
of Tropical Medicine of Rio Grande do Norte, Sen. Salgado Filho Av. 3000. Lagoa Nova, 59078970 Natal, RN, Brazil
| | - João Firmino Rodrigues Neto
- Institute
of Tropical Medicine of Rio Grande do Norte, Sen. Salgado Filho Av. 3000. Lagoa Nova, 59078970 Natal, RN, Brazil
- Multicamp
School of Medical Sciences, Federal University
of Rio Grande do Norte. Manuel Elpídio St. 610, Penedo, 59300000 Caicó, RN, Brazil
| | - Mary E. Wilson
- Departments
of Internal Medicine and Microbiology & Immunology, University of Iowa, 601, Hwy 6 West. 52242 Iowa City, Iowa, United States
- National
Institute of Science and Technology of Tropical Diseases, INCT-DT, 59078970 Natal, RN, Brazil
| | - Selma Maria Bezerra Jerônimo
- Institute
of Tropical Medicine of Rio Grande do Norte, Sen. Salgado Filho Av. 3000. Lagoa Nova, 59078970 Natal, RN, Brazil
- National
Institute of Science and Technology of Tropical Diseases, INCT-DT, 59078970 Natal, RN, Brazil
- Department
of Biochemistry, Federal University of Rio
Grande do Norte, Sen.
Salgado Filho Av., 3000, Lagoa Nova, 59078970 Natal, RN, Brazil
| |
Collapse
|
42
|
Aslanian-Kalkhoran L, Nouri N, Soltani-Zangbar MS, Mardi A, Aghebati-Maleki L. Immunoglobulin therapy for infertility and the role of immune cells in pregnancy success: An extensive investigation and update. J Reprod Immunol 2025; 169:104458. [PMID: 40015106 DOI: 10.1016/j.jri.2025.104458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
In the United States, roughly one out of every eight couples, or 7.5 million women, experience challenges related to conceiving or maintaining a pregnancy. The body's immune response is vital during pregnancy. T cells, natural killer (NK) cells, B cells, and macrophages (MQ) are immune cells in the female reproductive tract. They are in charge of maintaining tissue homeostasis and regulating the immune system's response to invasive pathogens. Failure to regulate these immune cells might result in inflammation, which reduces fertility. The immune system modulation of pregnancy loss has been studied with intralipid, intravenous immunoglobulin (IVIG), and paternal leukocyte vaccination. A concentrated antibody called intravenous immunoglobulin (IVIG) is utilized as a biological agent to treat autoimmune, viral, and inflammatory diseases and some immunodeficiencies. The main objective of this treatment is to restore a damaged immune system. IgGs, through binding to specific antigens, promote the innate immunity's cellular and humoral immune response by activating complements and binding to Fc receptors of several immune cells. Contrariwise, IVIG regulates pathogenic autoimmunity in animal models, including skin-blister diseases, nephrotoxic nephritis, and K/BxN arthritis. IVIG has, therefore, been of great interest as an immune modulator in several immune disorders. This review aims to investigate the immunological reasons of reproductive failure, focusing on the immunomodulatory effects of IVIG in its treatment.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narjes Nouri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
43
|
Kumar A, Johnson D, Bukowski A, Noto MJ, Carbonetti NH. Interferon lambda signaling in neutrophils enhances the pathogenesis of Bordetella pertussis infection. J Leukoc Biol 2025; 117:qiae202. [PMID: 39302155 PMCID: PMC11879762 DOI: 10.1093/jleuko/qiae202] [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: 06/06/2024] [Revised: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024] Open
Abstract
Interferon lambda plays diverse roles in bacterial infections. Previously, we showed that interferon lambda is induced in the lungs of Bordetella pertussis-infected adult mice and exacerbates inflammation. Here, we report that mice lacking the interferon lambda receptor 1 specifically on neutrophils (MRP8creIFNLR1fl/fl mice) exhibit reduced lung bacterial load and inflammation compared to wild-type mice during B. pertussis infection. In B. pertussis-infected wild-type mice, lung type I and III IFN responses were higher than in MRP8creIFNLR1fl/fl mice, correlating with increased lung inflammatory pathology. There was an increased proportion of interferon gamma-producing neutrophils in the lungs of MRP8creIFNLR1fl/fl mice compared to wild-type mice. IFNLR1-/- neutrophils incubated with B. pertussis exhibited higher killing compared to wild-type neutrophils. Treatment of wild-type neutrophils with interferon lambda further decreased their bacterial killing capacity and treatment of wild-type mice with interferon lambda increased lung bacterial loads. Contributing to the differential killing, we found that IFNLR1-/- neutrophils exhibit higher levels of reactive oxygen species, myeloperoxidase, matrix metalloproteinase-9 activity, neutrophil extracellular traps, and interferon gamma secretion than wild-type neutrophils, and inhibiting NADPH oxidase inhibited bacterial killing in IFNLR1-/- neutrophils. B. pertussis-induced interferon lambda secretion and IFNLR1 gene expression in mouse and human neutrophils and this was dependent on the bacterial virulence protein pertussis toxin. Pertussis toxin enhanced bacterial loads in wild type but not in MRP8creIFNLR1fl/fl or IFNLR1-/- mice. Thus, pertussis toxin disrupts neutrophil function by enhancing type III IFN signaling, which prevents neutrophils from effectively clearing B. pertussis during infection, leading to higher bacterial loads and exacerbation of lung inflammation.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Da’Kuawn Johnson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alicia Bukowski
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael J. Noto
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicholas H. Carbonetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
44
|
Silva LMR, López-Osorio S, Peixoto R, Zhou E, Espinosa G, Gärtner U, Taubert A, Conejeros I, Hermosilla C. Cellular immune responses of bovine polymorphonuclear neutrophils to Calicophoron daubneyi. Front Immunol 2025; 16:1515419. [PMID: 40018045 PMCID: PMC11865088 DOI: 10.3389/fimmu.2025.1515419] [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: 10/22/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Calicophoron daubneyi infections have increased in Europe, being more frequent than fasciolosis in some areas. Infection occurs once definitive hosts ingest encysted metacercariae present on vegetation. Following excystation, juvenile flukes penetrate the small intestinal mucosa and migrate into the rumen where adults mature. Throughout the somatic migration, juveniles come across different microenvironments and tissues and encounter host leukocytes. Besides phagocytosis, production of reactive oxygen species (ROS) and degranulation, polymorphonuclear neutrophils also cast neutrophil extracellular traps (NETs), which can entrap several parasite species, including the closely related liver fluke Fasciola hepatica. In this study, we analyzed whether in vitro exposure of bovine neutrophils to C. daubneyi antigen (CdAg) and eggs triggered neutrophils activation and NET formation. Results on scanning electron microscopy (SEM) and immunofluorescence analyses show weak formation of short spread NETs upon CdAg stimulation, corroborated by increased extracellular DNA measurements. Likewise, early NETosis was confirmed via nuclear area expansion assays. Bovine neutrophil stimulation with CdAg 100 µg/mL concentration led to a significant increase in oxygen consumption rates (p = 0.0152) and extracellular acidification rates (p = 0.0022), while lower concentrations of CdAg (10 µg/mL) failed to induce neutrophil activation, suggesting a dose dependent response. Both intra- and extracellular ROS production was not affected by any CdAg concentration here studied. Bovine neutrophil total adenosine triphosphate concentration significantly decreased after exposure to CdAg 100 µg/mL, in line to the observed with the positive control (phorbol myristate acetate/ionomycin). In summary, C. daubneyi activates bovine neutrophils with rather weak responses, which might suggest that the release of C. daubneyi-specific molecules (i.e. excretory-secretory antigens, proteases, or nucleases) could interfere with neutrophil-related effector mechanisms. Further ex vivo analyses will clarify if such mechanisms are also involved in pathogenesis of paramphistomosis by demonstrating neutrophil recruitment into affected intestinal mucosa.
Collapse
Affiliation(s)
- Liliana M. R. Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health and Science, Almada, Portugal
- Mediterranean Institute for Agriculture, Environment and Development (MED) and Global Change and Sustainability Institute (CHANGE), University of Évora, Évora, Portugal
| | - Sara López-Osorio
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
- CIBAV Research Group, Faculty of Agrarian Sciences, University of Antioquia, Medellín, Colombia
| | - Raquel Peixoto
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Ershun Zhou
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong, China
| | - Gabriel Espinosa
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Faculty of Human Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Iván Conejeros
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
45
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [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: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
46
|
Kim JY, Han HJ. Case report: In vivo detection of neutrophil extracellular traps in a dog with thrombosis induced by bacterial vasculitis. Front Vet Sci 2025; 12:1470605. [PMID: 40012748 PMCID: PMC11862914 DOI: 10.3389/fvets.2025.1470605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/10/2025] [Indexed: 02/28/2025] Open
Abstract
This case report describes NETosis as a cause of thrombosis in an 18.3 kg, 8-year-old intact male mixed-breed dog with bacterial vasculitis. The dog presented with sudden paresis of the thoracic limb, characterized by cyanosis, absent arterial pulse, and decreased peripheral blood glucose levels. Doppler ultrasound confirmed thrombosis in the dorsal common digital artery. Histopathology post-amputation revealed bacterial vasculitis, thrombosis, and infarction, with immunohistochemical staining identifying extracellular citrullinated histone H3 (CitH3), indicative of NETs involvement. Treatment included antibiotics, pentoxifylline, and anticoagulants, showing transient improvement before disease progression and euthanasia due to respiratory signs. These findings suggest NETs as a potential therapeutic target for bacterial vasculitis in similar cases.
Collapse
Affiliation(s)
| | - Hyun-Jung Han
- Department of Veterinary Emergency and Critical Care, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
47
|
Zhou Q, Zhou X, Li J, Wang R, Xie F. Research progress on the relationship between neutrophil extra-cellular traps and autogenous arteriovenous fistula thrombosis. J Vasc Access 2025:11297298251317298. [PMID: 39935409 DOI: 10.1177/11297298251317298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Autogenous arteriovenous fistula (AVF) is the preferred vascular access for long-term hemodialysis, and thrombosis is one of the most common complications. In recent years, it has been found that neutrophil extra-cellular traps (NETs) play an important role in thrombosis. NETs are a kind of network structure with DNA as a skeleton and intercalated with a variety of granule proteins, proteolytic enzymes, antimicrobial peptides and histone proteins, which are released into the extracellular space by neutrophils after stimulation. In this paper, the NETs in the role of AVF thrombus formation and NETs in the value of prevention and cure of AVF thrombus complications were reviewed.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xuhua Zhou
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junlin Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Runxiu Wang
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fuhua Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
48
|
Oh SA, Seol SI, Davaanyam D, Kim SW, Lee JK. Platelet-derived HMGB1 induces NETosis, exacerbating brain damage in the photothrombotic stroke model. Mol Med 2025; 31:46. [PMID: 39910417 PMCID: PMC11796003 DOI: 10.1186/s10020-025-01107-7] [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: 11/17/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Following cerebral ischemia, neutrophil extracellular traps (NETs) contribute significantly to brain damage by exacerbating delayed immune cell infiltration and vascular injury. They are detected both in brain tissue and within blood vessels. Danger-associated molecular pattern (DAMP) molecules have been implicated in inducing NETosis after cerebral ischemia. This study investigated the role of High mobility group box 1 (HMGB1), a prototype DAMP molecule, in NETosis induction following photothrombotic stroke (PTS), with a particular focus on neutrophil-platelet interactions. In PTS, thrombi consist primarily of aggregated platelets and neutrophils, lacking significant fibrin content. Triphenyltetrazolium chloride (TTC) staining revealed rapid but progressive expansion of the infarct area in the PTS model, commencing within 1 h and continuing until 24 h. Concomitant with this, peripheral neutrophils isolated following PTS exhibited progressive NETosis, particularly intravascular NETosis. This was evidenced by significant increase in citrullinated histone H3 (CitH3), a marker of NETosis, as early as 1 h post-PTS. Furthermore, serum levels of free DNA gradually and significantly increased, further supporting the induction of NETosis following PTS. Intranasal administration of BBCA, a peptidylarginine deiminase (PAD) inhibitor, effectively suppressed the induction of intravascular NETosis. Importantly, BBCA administration, both 30 min before and 4 h after PTS surgery, significantly reduced infarct volumes at 24 h and improved neurological outcomes. These findings underscore the crucial role of NETosis in both the initiation and progression of ischemic brain damage in this model. Following PTS, HMGB1 rapidly accumulated in serum, detectable as early as 1 h. Immunofluorescence staining revealed initial localization of HMGB1 in neurons, followed by its accumulation within activated neutrophils and platelets within blood vessels. Functional inhibition of HMGB1 by intranasal administration of an HMGB1 A box 4 h post-PTS significantly suppressed NETosis induction, reduced infarct volume, and improved neurological deficits, confirming the pivotal role of HMGB1 in NETosis induction. Notably, we observed a rapid platelet activation and concomitant HMGB1 induction within activated platelets after PTS. Co-culture experiments using naïve PMNs-platelets isolated following PTS demonstrate that extracellular HMGB1, particularly one derived from platelets, plays a critical role in activating neutrophils and inducing intravascular NETosis via a TLR4-dependent manner. Collectively, these findings highlight the critical role of NETosis not only in the initial stages of thrombus formation but also in the subsequent progression of ischemic brain damage in the PTS animal model. HMGB1, particularly platelet-derived HMGB1, emerges as a key mediator to this process. Therefore, targeting NETosis through modulation of HMGB1 presents a promising multipotent therapeutic strategy for mitigating ischemic brain damage.
Collapse
Affiliation(s)
- Sang-A Oh
- Department of Anatomy, Inha University School of Medicine, Iinha 100, Nam-Gu, Inchon, 22212, Republic of Korea
| | - Song-I Seol
- Department of Anatomy, Inha University School of Medicine, Iinha 100, Nam-Gu, Inchon, 22212, Republic of Korea
| | - Dashdulam Davaanyam
- Department of Anatomy, Inha University School of Medicine, Iinha 100, Nam-Gu, Inchon, 22212, Republic of Korea
| | - Seung-Woo Kim
- Department of Biomedical Sciences, Inha University School of Medicine, Inchon, Republic of Korea
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Iinha 100, Nam-Gu, Inchon, 22212, Republic of Korea.
| |
Collapse
|
49
|
Retter A, Singer M, Annane D. "The NET effect": Neutrophil extracellular traps-a potential key component of the dysregulated host immune response in sepsis. Crit Care 2025; 29:59. [PMID: 39905519 PMCID: PMC11796136 DOI: 10.1186/s13054-025-05283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs) as part of a healthy host immune response. NETs physically trap and kill pathogens as well as activating and facilitating crosstalk between immune cells and complement. Excessive or inadequately resolved NETs are implicated in the underlying pathophysiology of sepsis and other inflammatory diseases, including amplification of the inflammatory response and inducing thrombotic complications. Here, we review the growing evidence implicating neutrophils and NETs as central players in the dysregulated host immune response. We discuss potential strategies for modifying NETs to improve patient outcomes and the need for careful patient selection.
Collapse
Affiliation(s)
- Andrew Retter
- Critical Care, Guy's and St Thomas' NHS Foundation Trust, London, UK.
- School of Immunology and Microbial Sciences, King's College, London, UK.
- Volition, London, UK.
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- IHU PROMETHEUS, Comprehensive Sepsis Center, Garches, France
- University Versailles Saint Quentin-University Paris Saclay, INSERM, Garches, France
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), Garches, France
| |
Collapse
|
50
|
Ando S, Miyatake JI, Takeda M, Amakawa R, Nakamine H. Langerhans Cell Histiocytosis With Unusual Hexagonal Crystals in Addition to Usual Charcot-Leyden Crystals. Report of a Patient With Possible Process of Crystal Formation and Clinical Significance of a "Necrotic" Change. Int J Surg Pathol 2025; 33:153-159. [PMID: 38646671 DOI: 10.1177/10668969241246456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Langerhans cell histiocytosis is a rare neoplastic disorder characterized by the proliferation of Langerhans cells and often accompanied by eosinophil infiltration. Charcot-Leyden crystals, composed of galectin 10, are occasionally observed in Langerhans cell histiocytosis; however, histological images are rarely reported. We herein present a patient with Langerhans cell histiocytosis with Charcot-Leyden crystals and hexagonal crystals by describing the histologic and immunohistochemical features of a lymph node. A unique distribution of Charcot-Leyden crystals and hexagonal crystals was observed in this patient, shedding light on their possible formation process of the latter. We discuss the biological significance of eosinophilic abscesses in Langerhans cell histiocytosis and propose that these crystals may be linked to extracellular trap-cell death (ETosis). This example challenges the conventional characterization of "necrosis" in Langerhans cell histiocytosis and underscores the importance of recognizing ETosis as a potential mechanism involved in the pathogenesis of Langerhans cell histiocytosis. Further studies are underway to validate significance of these findings in a larger cohort of Langerhans cell histiocytosis patients.
Collapse
Affiliation(s)
- Sayaka Ando
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | | | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | - Ryuichi Amakawa
- Department of Hematology, Japan Baptist Hospital, Kyoto, Japan
| | - Hirokazu Nakamine
- Department of Pathology and Laboratory Medicine, Japan Baptist Hospital, Kyoto, Japan
| |
Collapse
|