1
|
Zhang Z, Wang Y, Li T, Wang H. NETosis in myocardial ischemia-reperfusion injury: From mechanisms to therapies (Review). Biomed Rep 2025; 23:113. [PMID: 40420974 PMCID: PMC12105085 DOI: 10.3892/br.2025.1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
The present review describes the mechanisms of NETosis and its role in myocardial ischemia-reperfusion injury (MIRI), focusing on the release of neutrophil extracellular traps (NETs) by activated neutrophils. NETs, composed of depolymerized chromatin and granule proteins, are crucial for pathogen entrapment, infection control and immune regulation. However, NET formation, linked to neutrophil death (NETosis), exacerbates MIRI by promoting inflammation and tissue damage. To address therapeutic strategies for NETosis in MIRI, several potential clinically significant approaches were explored, including peptidylarginine deaminase 4 inhibition, DNase therapy, antioxidants, inflammation modulation, and antithrombotic treatments, which not only provide novel diagnostic biomarkers and therapeutic targets in MIRI, but are also expected to improve patient prognosis and advance the development of personalised medicine.
Collapse
Affiliation(s)
- Ziyang Zhang
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanxin Wang
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Tie Li
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Hongfeng Wang
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
2
|
Lee JY, Cho HR, Oh HG, Hwang JH. Comparative transcriptome analysis of PBMCs in cats diagnosed with and recovered from FIPV. Lab Anim Res 2025; 41:18. [PMID: 40514753 DOI: 10.1186/s42826-025-00247-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/27/2025] [Accepted: 05/12/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Feline infectious peritonitis is a viral disease caused by feline coronavirus an enveloped virus with a single-stranded RNA genome that is approximately 30 kb long. Although FCoV generally causes mild symptoms, approximately 5% of cases progress to death in cats worldwide. FCoV shares certain virological features with severe acute respiratory syndrome coronavirus 2 that causes COVID-19, indicating that common therapeutic strategies may be applicable. GS-441524 the parent drug of remdesivir and a competitive inhibitor of nucleoside triphosphates in viral RNA synthesis is a well-known treatment for FIP. However, comparative transcriptome and gene ontology analyses of normal (Normal), FIP-diseased (FIPD), and FIP-recovered (FIPR) cats have not yet been conducted. RESULTS In this study, we compared the mRNA expression profiles of peripheral blood mononuclear cells from Normal, FIPD, and FIPR cats to identify immunological alterations. We identified 677 (FIPD/Normal) and 431 (FIPR/FIPD) differentially expressed genes with statistical significance. These data were input into the bioinformatics program. As a result, the analysis revealed statistically significant and contrasting patterns of canonical pathways of neutrophil degranulation and interleukin-8 (IL-8) signaling pathways. Additionally, we observed that kruppel-like factor 6 (KLF6) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) were upstream molecules of IL-8, promoting neutrophil activation and function. CONCLUSIONS This study identified immunological alterations in PBMCs of Normal, FIPD, and FIPR cats. KLF-6 and NF-κB were found to regulate IL-8-mediated neutrophil activation.
Collapse
Affiliation(s)
- Ju Young Lee
- Center for Large Animals Convergence Research, Korea Institue of Toxicology, 30 Baekhak1- gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea
| | - Hyeong Ryeol Cho
- Center for Large Animals Convergence Research, Korea Institue of Toxicology, 30 Baekhak1- gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea
| | - Hong-Geun Oh
- Center for Companion Animal New Drug Development, Korea Institue of Toxicology, 30 Baekhak1-gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea
- R&D Division, HUVET Co., Ltd, Hanaro, Iksan-si, Jellabuk-do, 483-26, Republic of Korea
| | - Jeong Ho Hwang
- Center for Large Animals Convergence Research, Korea Institue of Toxicology, 30 Baekhak1- gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea.
- Center for Companion Animal New Drug Development, Korea Institue of Toxicology, 30 Baekhak1-gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea.
- Center for Animal Medicine and Foods, Korea Institue of Toxicology, 30 Baekhak1-gil, Jeongeup, Jellabuk-do, 56212, Republic of Korea.
| |
Collapse
|
3
|
Kindberg KM, Nordeng J, Langseth MS, Schandiz H, Roald B, Solheim S, Seljeflot I, Stokke MK, Helseth R. IL-6R Signaling Is Associated with PAD4 and Neutrophil Extracellular Trap Formation in Patients with STEMI. Int J Mol Sci 2025; 26:5348. [PMID: 40508157 PMCID: PMC12154504 DOI: 10.3390/ijms26115348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/27/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Inflammation contributes to myocardial injury in ST-elevation myocardial infarction (STEMI). Interleukin-6 receptor (IL-6R) inhibition has been shown to mitigate myocardial injury and reduce levels of the prothrombotic and inflammatory mediator, neutrophil extracellular traps (NETs). The enzyme peptidylarginine deiminase 4 (PAD4) is central in NET formation. We hypothesized that PAD4 links IL-6R activation and NET formation. METHODS We conducted thrombus aspiration and peripheral blood sampling in 33 STEMI patients. In thrombi and leukocytes, we quantified the mRNA of IL-6, IL-6R, and PAD4. In peripheral blood, the protein levels of IL-6, IL-6R, PAD4, dsDNA, H3Cit, MPO-DNA, and troponin T were quantified. RESULTS In thrombi and circulating leukocytes, PAD4 mRNA was associated with IL-6R mRNA (thrombi: β = 0.34, 95% CI [0.16-0.53], p = 0.001, circulating leukocytes: β = 0.92, 95% CI [0.07-1.77], p = 0.036). There were no correlations between PAD4 and IL-6 in thrombi and leukocytes. The protein levels of IL-6R were associated with the NET marker H3Cit (rs = 0.40, p = 0.02). In thrombi, PAD4 mRNA was associated with high levels of troponin T (β = 1.15 95% CI [0.27-2.04], p = 0.013). CONCLUSION We demonstrate an association between PAD4, IL-6R, and troponin release in STEMI patients. Our findings indicate a PAD4-mediated connection between IL-6R and NET formation and highlight PAD4 as a potential treatment target for mitigating inflammation and myocardial injury in STEMI.
Collapse
Affiliation(s)
- Kristine Mørk Kindberg
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, 0450 Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Jostein Nordeng
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, 0450 Oslo, Norway
| | | | - Hossein Schandiz
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
- Department of Oncology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Borghild Roald
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
- Department of Pathology, Oslo University Hospital Ullevaal, 0450 Oslo, Norway
| | - Svein Solheim
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, 0450 Oslo, Norway
| | - Ingebjørg Seljeflot
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, 0450 Oslo, Norway
| | - Mathis Korseberg Stokke
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - Ragnhild Helseth
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, 0450 Oslo, Norway
| |
Collapse
|
4
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [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: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
5
|
Nakazawa D, Masuda S, Nishibata Y, Watanabe-Kusunoki K, Tomaru U, Ishizu A. Neutrophils and NETs in kidney disease. Nat Rev Nephrol 2025; 21:383-398. [PMID: 40102634 DOI: 10.1038/s41581-025-00944-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
6
|
Zambrano F, Uribe P, Schulz M, Hermosilla C, Taubert A, Sánchez R. Antioxidants as Modulators of NETosis: Mechanisms, Evidence, and Therapeutic Potential. Int J Mol Sci 2025; 26:5272. [PMID: 40508099 PMCID: PMC12154535 DOI: 10.3390/ijms26115272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/22/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Neutrophil extracellular trap (NET) formation is a process known as NETosis and is a critical innate immune response mechanism that can become pathologically dysregulated in various inflammatory, autoimmune, infectious, and neoplastic diseases. Reactive oxygen species (ROS) play a central role in NETosis induction, making antioxidants a promising therapeutic approach. This review outlines the molecular mechanisms underlying NET formation and highlights three principal antioxidant-based inhibitory strategies: NADPH oxidase (NOX) inhibition, ROS scavenging, and myeloperoxidase (MPO) inhibition. Evidence supports the use of agents such as diphenylene iodonium (NOX inhibitor), N-acetylcysteine and glutathione (ROS scavengers), and thiocyanate (MPO inhibitor), which significantly reduce NETosis in vitro and in vivo. Moreover, natural compounds like resveratrol show pleiotropic effects by modulating neutrophil activation, ROS production, and protease activity. Combination therapies that enhance total antioxidant capacity are particularly effective, though their translation to clinical practice faces challenges such as stimulus specificity, bioavailability, and maintaining immune competence. Antioxidant-based therapies thus represent a promising avenue for targeted NETosis modulation. Future research should focus on improving delivery systems, identifying NET-specific biomarkers, and integrating antioxidants into broader immunomodulatory strategies.
Collapse
Affiliation(s)
- Fabiola Zambrano
- Center of Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT—BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (F.Z.); (P.U.); (M.S.)
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Pamela Uribe
- Center of Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT—BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (F.Z.); (P.U.); (M.S.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Mabel Schulz
- Center of Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT—BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (F.Z.); (P.U.); (M.S.)
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (C.H.); (A.T.)
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (C.H.); (A.T.)
| | - Raúl Sánchez
- Center of Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT—BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile; (F.Z.); (P.U.); (M.S.)
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
7
|
Wang Y, Liu Y, Du C, Zhang Y, Zhu L. Mouse Bone Marrow Neutrophil Extracellular Trap Proteomics by Microbial Stimuli. Sci Data 2025; 12:853. [PMID: 40410159 PMCID: PMC12102246 DOI: 10.1038/s41597-025-05181-8] [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: 02/18/2025] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
Neutrophils, the most abundant white blood cells in human circulation, play a crucial role in innate immunity. One of their key defense mechanisms is the formation of neutrophil extracellular traps (NETs), web-like structures composed of chromatin and antimicrobial proteins that help capture and neutralize pathogens. While previous studies have identified a limited set of NET-associated proteins, the comprehensive proteomic landscape of NETs induced by different stimuli remains largely unexplored. In this study, we used data-independent acquisition mass spectrometry to analyze the proteomic composition of NETs induced by five distinct stimuli: β-glucan, lipopolysaccharide, polyinosinic-polycytidylic acid sodium, resiquimod, and severe fever with thrombocytopenia syndrome bunyavirus. Across all conditions, we identified 5,868 NET-associated proteins, revealing significant stimulus-dependent differences in protein composition. Notably, differentially expressed proteins were detected in each condition, highlighting unique proteomic signatures that may reflect distinct immune responses. This dataset offers key insights into the proteomic diversity of NETs and their role in immune regulation, providing a foundation for future research on NET-mediated immunity in infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yijie Wang
- Beijing Key Laboratory of Viral Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yujia Liu
- Beijing Key Laboratory of Viral Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chunjing Du
- Beijing Key Laboratory of Viral Infectious Diseases, Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yue Zhang
- Beijing Key Laboratory of Viral Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
- Beijing Institute of Infectious Diseases, Beijing, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| | - Liuluan Zhu
- Beijing Key Laboratory of Viral Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
- Beijing Institute of Infectious Diseases, Beijing, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Arya SB, Collie SP, Xu Y, Fernandez M, Sexton JZ, Mosalaganti S, Coulombe PA, Parent CA. Neutrophils secrete exosome-associated DNA to resolve sterile acute inflammation. Nat Cell Biol 2025:10.1038/s41556-025-01671-4. [PMID: 40404894 DOI: 10.1038/s41556-025-01671-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 04/09/2025] [Indexed: 05/24/2025]
Abstract
Acute inflammation, characterized by a rapid influx of neutrophils, is a protective response that can lead to chronic inflammatory diseases when left unresolved. We previously showed that secretion of LTB4-containing exosomes via nuclear envelope-derived multivesicular bodies is required for effective neutrophil infiltration during inflammation. Here we report that the co-secretion of these exosomes with nuclear DNA facilitates the resolution of the neutrophil infiltrate in a mouse skin model of sterile inflammation. Activated neutrophils exhibit rapid and repetitive DNA secretion as they migrate directionally using a mechanism distinct from suicidal neutrophil extracellular trap release and cell death. Packaging of DNA in the lumen of nuclear envelope-multivesicular bodies is mediated by lamin B receptor and chromatin decondensation. These findings advance our understanding of neutrophil functions during inflammation and the physiological relevance of DNA secretion.
Collapse
Affiliation(s)
- Subhash B Arya
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Samuel P Collie
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yang Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin Fernandez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Shyamal Mosalaganti
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carole A Parent
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [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: 08/30/2024] [Revised: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
10
|
Katayama H. Neutrophil Extracellular Traps Capturing SARS-CoV-2 in the Lung Tissue (Alveoli and Parenchyma) Cause Microthrombi - A Strategy to Eliminate SARS-CoV-2 From the Circulation as Degraded Fibrin Clots. Circ Rep 2025; 7:379-382. [PMID: 40352121 PMCID: PMC12061506 DOI: 10.1253/circrep.cr-24-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 05/14/2025] Open
Abstract
Background It has been thought that neutrophil extracellular traps (NETs) and thrombosis exacerbate COVID-19, but, on the other hand, NETs are an important player in innate immunity. The precise roles of NETs and thrombosis in the course of COVID-19 have not been fully elucidated. Methods and Results The roles were investigated in the literature and a new theory was formulated. When neutrophils encounter SARS-CoV-2 in the lung tissue, they undergo NETosis and capture the virus. This capture is triggered by electrostatic interaction between histones in NETs and SARS-CoV-2; histones are highly positively charged, and viruses, including SARS-CoV-2, have a net negative charge under physiological pH. NETs that capture SARS-CoV-2 fall into alveolar capillaries through the collapsed endothelium to spare the lung tissue from the toxicity of NETs. NETs in the microvessels cause microthrombosis; positively charged histones induce the aggregation of negatively charged platelets, which leads to microthrombi. Microthrombi engulfing SARS-CoV-2 are consolidated into fibrin clots, which are eventually degraded by increased fibrinolysis and eliminated from the circulation. Conclusions This novel theory suggests that NETosis and microthrombosis are phenomena inevitably elicited in COVID-19, and in combination they are a system newly termed "NETombosis". Undegraded fibrin clots remaining in the microcirculation may be the cause of the sequelae, because they cause long-lasting circulatory failure in various organs.
Collapse
|
11
|
Zeng M, Niu Y, Huang J, Deng L. Advances in neutrophil extracellular traps and ferroptosis in sepsis-induced cardiomyopathy. Front Immunol 2025; 16:1590313. [PMID: 40356926 PMCID: PMC12066755 DOI: 10.3389/fimmu.2025.1590313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Sepsis-induced cardiomyopathy is a reversible non-ischemic acute cardiac dysfunction associated with sepsis. It is strongly associated with an abnormal immune response. It emerges as a vital threat to public health owing to its high mortality rate. However, the exact pathogenesis requires further investigation. In recent years, NETosis and ferroptosis, which are novel modes of programmed cell death, have been identified and found to play important roles in sepsis-related organ damage. This article outlines the mechanisms of these two modes of cell death, discusses the role of neutrophil extracellular traps in myocardial injury and the importance of ferroptosis in sepsis-induced cardiomyopathy, and reviews the potential interconnection between these two types of programmed cell death in sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
| | | | | | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical
University, Zhanjiang, China
| |
Collapse
|
12
|
Sugimoto K, Yang C, Ono M, Shirazaki M, Katada R, Matsumoto H. Neutrophils induce astrocytic AQP4 expression via IL-1α and TNF, contributing to cerebral oedema in ischaemic stroke rats. Sci Rep 2025; 15:13923. [PMID: 40263535 PMCID: PMC12015259 DOI: 10.1038/s41598-025-98758-7] [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: 12/04/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
During the acute phase (1-3 days) of cerebral infarction, neutrophils and macrophages accumulate at the infarction site, inducing inflammation and cerebral oedema. However, the role of neutrophils in oedema formation after ischaemic stroke remains unclear. This study examined neutrophil involvement in cerebral oedema using a transient middle cerebral artery occlusion (tMCAO) rat model, primary cultured neutrophils, and astrocytes. Brain specimens were stained with myeloperoxidase (MPO) and lymphocyte antigen 6 complexes, locus G (Ly6G), and the number of MPO+/Ly6G+ cells was counted. Neutrophil infiltration began in the leptomeninges at 3 h, reaching the ischaemic cortex by 6 h and the striatum by 24 h, peaking at 24-48 h before declining. Neutrophils attached to endothelial walls and infiltrated the brain parenchyma, correlating with oedema severity. Infiltrating neutrophils strongly expressed IL-1α and TNF in the ischaemic brain. Co-culturing LPS-activated neutrophils with astrocytes increased Aqp4 mRNA and protein expression, which was inhibited by IL-1RI and TNF antagonists. These findings suggest that activated neutrophils exacerbate cerebral oedema by inducing astrocytic AQP4 expression via IL-1α and TNF in peri-infarct and ischaemic core tissues.
Collapse
Affiliation(s)
- Kana Sugimoto
- Department of Legal Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Chihpin Yang
- Department of Legal Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Miharu Ono
- Department of Legal Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mai Shirazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Ryuichi Katada
- Department of Legal Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Division of Legal Medicine, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Hiroshi Matsumoto
- Department of Legal Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Education and Research Institute for Death Control and Prevention, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
13
|
Zhang Y, Si W, Mao Y, Xu S, Li F, Liu J, Du S, Shao J, Qi Y, Peng X, Xue M, Jiang M, Guo K, Hu Y, Zhang F. Upregulation of ferroptosis in glucocorticoids-induced posterior subcapsular cataracts. Commun Biol 2025; 8:613. [PMID: 40234585 PMCID: PMC12000516 DOI: 10.1038/s42003-025-08067-y] [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: 08/26/2024] [Accepted: 04/09/2025] [Indexed: 04/17/2025] Open
Abstract
The Glucocorticoid-induced posterior subcapsular cataracts (GIC) is a common complication of patients received glucocorticoid treatment in clinic. We find that dexamethasone (DEX) induces lens epithelial cells' ferroptosis. DEX treatment increases intracellular ferroptosis signatures in lens epithelial cell line in vitro as well as in rat lens in vivo. The inhibition of ferroptosis by liproxstatin-1 reduces the incidence of DEX-induced rat GIC. Experimental evidence and expression profiling showed that DEX induces ferroptosis through upregulating tetraspanin CD82- controlled P53 expression. DEX-activated glucocorticoid receptors directly bind to the CD82 promoter, driving its transcriptional upregulation. CD82 expression is upregulated in the anterior capsular epithelium of GIC patients as well as in the DEX-treated rat lens and caused the cell death of anterior capsule. DEX treatment and Overexpression of CD82 in cells recapitulated ferroptotic signatures through P53 activation and GPX4/SLC7A11 suppression. Taken together, GIC is closely associated with the upregulation of CD82-P53-GPX4/SLC7A11 axis-mediated ferroptosis.
Collapse
Affiliation(s)
- Yuhang Zhang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Wei Si
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yi Mao
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Su Xu
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fuzhen Li
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shanshan Du
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jingzhi Shao
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Ying Qi
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xuyan Peng
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mengjiao Xue
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mingjun Jiang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Keyu Guo
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yanzhong Hu
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- The Jointed National Laboratory of Antibody Engineering, Henan University, Kaifeng, China.
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| | - Fengyan Zhang
- The Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
14
|
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
|
15
|
Li X, Wu F, Yu D, Su X, Wang K, Huang Z, Lu Z. Archaea-inspired deoxyribonuclease I liposomes prevent multiple organ dysfunction in sepsis. J Control Release 2025; 380:1109-1126. [PMID: 39986474 DOI: 10.1016/j.jconrel.2025.02.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Neutrophil extracellular traps (NETs) and circulating cell-free DNA (cfDNA) are pivotal in driving excessive inflammation and organ damage during sepsis, with their levels correlating positively with sepsis severity in both patients and murine models. Despite the ability of deoxyribonuclease I (DNase I) to degrade NETs and cfDNA, its short half-life and rapid degradation limit its therapeutic effectiveness. To address this challenge, we developed a methyl-branched liposome fused with a red blood cell membrane for the systemic delivery of DNase I (DNase I/Rm-Lipo). The efficacy of DNase I/Rm-Lipo was evaluated in the stimulated immune cells and septic model. The data confirmed that DNase I/Rm-Lipo efficiently removed excess NETs and cfDNA in activated neutrophils. Following injection, DNase I/Rm-Lipo exhibited an extended circulation time, effectively suppressing neutrophil activation and regulating macrophage polarization to mitigate inflammation and prevent organ dysfunction in septic mice. These findings highlight the therapeutic potential of DNase I/Rm-Lipo as a promising candidate for sepsis management by targeting the degradation of NETs and cfDNA.
Collapse
Affiliation(s)
- Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Fan Wu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Dedong Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiayi Su
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Kaikai Wang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China
| | - Zhiwei Huang
- Central Laboratory, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui 323000, China; Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou 325035, China.
| |
Collapse
|
16
|
Shi G, Cao Y, Xu J, Chen B, Zhang X, Zhu Y, Liu L, Liu X, Zhang L, Zhou Y, Li S, Yang G, Liu X, Chen F, Chen X, Zhang J, Zhang S. Inhibition of S100A8/A9 ameliorates neuroinflammation by blocking NET formation following traumatic brain injury. Redox Biol 2025; 81:103532. [PMID: 39929053 PMCID: PMC11849670 DOI: 10.1016/j.redox.2025.103532] [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: 12/13/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Traumatic brain injury (TBI) triggers a robust inflammatory response that is closely linked to worsened clinical outcomes. S100A8/A9, also known as calprotectin or myeloid-related protein-8/14 (MRP8/14), is an alarmin primarily secreted by activated neutrophils with potent pro-inflammatory property. In this study, we explored the roles of S100A8/A9 in modulating neuroinflammation and influencing TBI outcomes, delving into the underlying mechanisms. S100A8/A9-enriched neutrophils were present in the injured brain tissue of TBI patients, and elevated plasma levels of S100A8/A9 were correlated with poorer neurological function. Furthermore, using a TBI mouse model, we demonstrated that treatment with the selective S100A8/A9 inhibitor Paquinimod significantly mitigated neuroinflammation and neuronal death, thereby improving the prognosis of TBI mice. Mechanistically, we found that S100A8/A9, in conjunction with neutrophil activation and infiltration into the brain, enhances reactive oxygen species (ROS) production within neutrophils, accelerating PAD4-mediated neutrophil extracellular trap (NET) formation, which in turn exacerbates neuroinflammation. These findings suggest that S100A8/A9 amplifies neuroinflammatory responses by promoting NET formation in neutrophils. Inhibition of S100A8/A9 effectively attenuated NET-mediated neuroinflammation; however, when PAD4 was overexpressed in the brain using adenovirus, leading to an increased formation of NET in the brain, the anti-inflammatory effects of S100A8/A9 inhibition were markedly diminished. Further experiments with PAD4 knockout mice confirmed that the reduction of NETs could substantially alleviate S100A8/A9-driven neuroinflammation. Finally, we established that the suppression of NET formation by S100A8/A9 inhibition is primarily mediated through the AMPK/Nrf2/HO-1 signaling pathway. These findings underscore the critical pathological role of S100A8/A9 in TBI and emphasize the need for further exploration of S100A8/A9 inhibitor Paquinimod as a potential therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Guihong Shi
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Jianye Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Bo Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xu Zhang
- School of Medicine, Nankai University, Tianjin, 300052, China
| | - Yanlin Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Liang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xilei Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Luyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Shenghui Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Guili Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xiao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China.
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China.
| |
Collapse
|
17
|
Zhou E, Li Y, Wu Z, Chen Y, Wu H, Ye Y, Li T, Wang J, Yang Z. Neutrophil extracellular traps formation and autophagy in bongkrekic acid exposed human neutrophils. Toxicol In Vitro 2025; 104:106003. [PMID: 39730015 DOI: 10.1016/j.tiv.2024.106003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024]
Abstract
Bongkrekic acid (BKA), a less well-known foodborne toxin, has been implicated in numerous poisoning incidents. Recent studies suggest that BKA exerts an impact on the immune system, particularly on innate immunity. The release of neutrophil extracellular traps (NETs) is relatively a newly-discovered mechanism involving innate immunity. This study was designed to characterize and evaluate the effects of BKA on human NET formation. The co-localization of DNA, histones, and myeloperoxidase (MPO) was determined via immunostaining to confirm BKA-triggered NET formation in human neutrophils. NET quantification showed that NET formation induced by BKA was both time- and dose-dependent, and was associated with p38, ERK, PAD4 and P2X1 receptor. Moreover, immunostaining analysis observed that BKA triggered both NET formation and autophagy. Additionally, pharmacological experiments revealed that autophagy mediated BKA-triggered NET formation. Collectively, these insights offer a novel perspective on the effects of BKA exposure on host's innate immune response, and may shed new light on BKA poisoning. We call for further work to be conducted in this field to unravel the intricate mechanisms governing NET formation and autophagy in the context of BKA poisoning.
Collapse
Affiliation(s)
- Ershun Zhou
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yifei Li
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhikai Wu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yichun Chen
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Hanpeng Wu
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yingrong Ye
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Tianyu Li
- Guangxi University, Nanning 530004, Guangxi Province, PR China
| | - Jingjing Wang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China.
| | - Zhengtao Yang
- School of Animal Science and Technology, Foshan University, Foshan 528225, Guangdong Province, PR China.
| |
Collapse
|
18
|
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
|
19
|
Li Y, Ren S, Zhou S. Advances in sepsis research: Insights into signaling pathways, organ failure, and emerging intervention strategies. Exp Mol Pathol 2025; 142:104963. [PMID: 40139086 DOI: 10.1016/j.yexmp.2025.104963] [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: 12/30/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Sepsis is a complex syndrome resulting from an aberrant host response to infection. A hallmark of sepsis is the failure of the immune system to restore balance, characterized by hyperinflammation or immunosuppression. However, the net effect of immune system imbalance and the clinical manifestations are highly heterogeneous among patients. In recent years, research interest has shifted from focusing on the pathogenicity of microorganisms to the molecular mechanisms of host responses which is also associated with biomarkers that can help early diagnose sepsis and guide treatment decisions. Despite significant advancements in medical science, sepsis remains a major challenge in healthcare, contributing to substantial morbidity and mortality worldwide. Further research is needed to improve our understanding of this condition and develop novel therapies to improve outcomes for patients with sepsis. This review explores the related signal pathways of sepsis and underscores recent advancements in understanding its mechanisms. Exploration of diverse biomarkers and the emerging concept of sepsis endotypes offer promising avenues for precision therapy in the future.
Collapse
Affiliation(s)
- Yehua Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China.
| | - Siying Ren
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Shen'ao Zhou
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
20
|
Cai Y, Yang Q, Tang X, Wang P, Cui J, Du X, Zhang T, Chen Y. Baicalin mitigates hyperglycemia-linked intestinal epithelial barrier impairment in part by inhibiting the formation of neutrophil extracellular traps. Front Immunol 2025; 16:1551256. [PMID: 40098957 PMCID: PMC11911346 DOI: 10.3389/fimmu.2025.1551256] [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: 12/25/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Background Under hyperglycemic conditions, impaired intestinal barrier integrity leads to heightened level of inflammation, playing important roles in driving diabetic complications. Emerging evidence supports the implications of neutrophil extracellular traps (NETs) in the pathogenesis of diabetes. However, whether NETs contribute to hyperglycemia-linked intestinal barrier impairment remains to be investigated. Moreover, baicalin, the major chemical component of Scutellaria baicalensis Georgi, is equipped with twofold intestinal protective and neutrophil suppressive activities. Yet, it is unclear if baicalin is effective at mitigating hyperglycemia-linked NETs-mediated intestinal barrier impairment. Methods To directly address the mechanistic implications of NETs in hyperglycemia-linked intestinal epithelial barrier impairment, the impact of DNase I treatment or Padi4 gene deficiency on intestinal epithelial integrity was first examined in the streptozotocin (STZ)-induced hyperglycemic mice in vivo. Next, the pharmacological impact of baicalin on NETs formation and intestinal epithelial barrier impairment was investigated in high glucose- and/or lipopolysaccharides (LPS)-stimulated neutrophils in vitro and in STZ-induced hyperglycemic mice in vivo, respectively. Results The in vitro experiments confirmed that high glucose and/or LPS induced NETs formation. NETs directly impaired the viability and tight junction of the intestinal epithelial cells. The histological and immunohistochemical examinations unveiled that along with impaired intestinal epithelial morphology, citrullinated histone H3 (H3Cit), a marker of NETs, and neutrophil specific Ly6G were readily detected in the intestinal epithelium in the hyperglycemic mice. Without affecting the presence of neutrophils, DNase I treatment or Padi4 gene deficiency markedly mitigated intestinal NETs formation and improved the intestinal morphology in the hyperglycemic mice. Notably, baicalin suppressed NETs formation and inhibited histone H3 citrullination stimulated by high glucose, LPS or both in vitro. Furthermore, baicalin blunted NETs formation and partially preserved the integrity of the intestinal epithelium in the hyperglycemic mice in vivo. Conclusions The current study sheds new light on the pathophysiological implications of NETs in intestinal epithelial barrier impairment under hyperglycemic conditions. Most importantly, the findings here demonstrate for the first time that baicalin directly inhibits NETs formation stimulated by high glucose and/or LPS, which may in part account for its pharmacological effects at protecting against hyperglycemia-linked intestinal epithelial barrier impairment.
Collapse
Affiliation(s)
- Yiqing Cai
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinbo Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xinmiao Tang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peiwei Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jingang Cui
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoye Du
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Zhang J, Shao Y, Wu J, Zhang J, Xiong X, Mao J, Wei Y, Miao C, Zhang H. Dysregulation of neutrophil in sepsis: recent insights and advances. Cell Commun Signal 2025; 23:87. [PMID: 39953528 PMCID: PMC11827254 DOI: 10.1186/s12964-025-02098-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/09/2025] [Indexed: 02/17/2025] Open
Abstract
Sepsis remains the leading cause of death in intensive care units. Despite newer antimicrobial and supportive therapies, specific treatments are still lacking. Neutrophils are pivotal components of the effector phase of the host immune defense against pathogens and play a crucial role in the control of infections under normal circumstances. In addition to its anti-infective effects, the dysregulation and overactivation of neutrophils may lead to severe inflammation or tissue damage and are potential mechanisms for poor prognosis in sepsis. This review focuses on recent advancements in the understanding of the functional status of neutrophils across various pathological stages of sepsis to explore the mechanisms by which neutrophils participate in sepsis progression and provide insights for the treatment of sepsis by targeting neutrophils.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingyi Wu
- Department of Anesthesiology, Zhongshan Hospital(Xiamen), Fudan University, Xiamen, China
| | - Jing Zhang
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, China
| | - Xiangsheng Xiong
- Department of Anesthesiology, Huai'an hospital affiliated to Yangzhou University (The fifth People's Hospital of Huai'an), Huai'an, Jiangsu, China
| | - Jingjing Mao
- Department of Anesthesiology, Huai'an hospital affiliated to Yangzhou University (The fifth People's Hospital of Huai'an), Huai'an, Jiangsu, China
| | - Yunwei Wei
- Department of Anesthesiology, Women's Health Center of Shanxi, Children's Hospital of Shanxi, Taiyuan, Shanxi, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Li H, Li C, Fu C, Wang Y, Liang T, Wu H, Wu C, Wang C, Sun T, Liu S. Innovative nanoparticle-based approaches for modulating neutrophil extracellular traps in diseases: from mechanisms to therapeutics. J Nanobiotechnology 2025; 23:88. [PMID: 39915767 PMCID: PMC11800495 DOI: 10.1186/s12951-025-03195-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/02/2025] [Indexed: 02/11/2025] Open
Abstract
Neutrophil extracellular traps (NETs) participate in both host defense and the pathogenesis of various diseases, such as infections, thrombosis, and tumors. While they help capture and eliminate pathogens, NETs' excessive or dysregulated formation can lead to tissue damage and disease progression. Therapeutic strategies targeting NET modulation have shown potential, but challenges remain, particularly in achieving precise drug delivery and maintaining drug stability. Nanoparticle (NP)-based drug delivery systems offer innovative solutions for overcoming the limitations of conventional therapies. This review explores the biological mechanisms of NET formation, their interactions with NPs, and the therapeutic applications of NP-based drug delivery systems for modulating NETs. We discuss how NPs can be designed to either promote or inhibit NET formation and provide a comprehensive analysis of their potential in treating NET-related diseases. Additionally, we address the current challenges and future prospects for NP-based therapies in NET research, aiming to bridge the gap between nanotechnology and NET modulation for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Can Li
- Department of Hematology, The Second Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chenxi Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
25
|
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
|
26
|
Shim HB, Deniset JF, Kubes P. Knowing when to stop: MICL self-regulates neutrophil NETosis. Cell Res 2025; 35:89-90. [PMID: 39402399 PMCID: PMC11770063 DOI: 10.1038/s41422-024-01037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Affiliation(s)
- Hanjoo Brian Shim
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Justin François Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
- Department of Biomedical and Molecular Science, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
27
|
Manoj H, Gomes SM, Thimmappa PY, Nagareddy PR, Jamora C, Joshi MB. Cytokine signalling in formation of neutrophil extracellular traps: Implications for health and diseases. Cytokine Growth Factor Rev 2025; 81:27-39. [PMID: 39681501 DOI: 10.1016/j.cytogfr.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Neutrophils, as essential component of the innate immune response, form a crucial part in the defence mechanisms through the release of extracellular traps (NETs). These web-like structures, composed of chromatin and antimicrobial proteins, are essential for the entrapment and inactivation of pathogens. However, either constitutive formation or inefficient clearance of NETs leads to adverse effects such as fibrosis, thrombosis, delayed wound healing and tissue damage in multiple diseases associated with sterile inflammation. This dichotomy casts NETs as both protective agents and harmful factors in several diseases such as autoimmune diseases, metabolic syndromes, systemic infections, and malignancies. Besides microbes and their products, variety of stimulants including pro-inflammatory cytokines induce NETs. The complex interactions and cross talk among the pro-inflammatory cytokines including IL-8, IL-6, GM-CSF, TNF-α, IFNs, and IL-1β activate neutrophils to form NETs and also contributes to a vicious circle of inflammatory cascade, leading to increased inflammation, oxidative stress, and thrombotic events. Emerging evidence indicates that the dysregulated cytokine milieus in diseases, such as diabetes mellitus, obesity, atherosclerosis, stroke, rheumatoid arthritis, and systemic lupus erythematosus, potentiate NETs release, thereby promoting disease development. Thus, neutrophils represent both critical effectors and potential therapeutic targets, underscoring their importance in the context of cytokine-mediated therapies for a spectrum of diseases. In the present review, we describe various cytokines and associated signalling pathways activating NETs formation in different human pathologies. Further, the review identifies potential strategies to pharmacologically modulate cytokine pathways to reduce NETs.
Collapse
Affiliation(s)
- Haritha Manoj
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sarah Michael Gomes
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma, OK, USA
| | - Colin Jamora
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh 201314, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
28
|
Liu X, Zheng Y, Meng Z, Wang H, Zhang Y, Xue D. Gene Regulation of Neutrophils Mediated Liver and Lung Injury through NETosis in Acute Pancreatitis. Inflammation 2025; 48:393-411. [PMID: 38884700 DOI: 10.1007/s10753-024-02071-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024]
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal emergencies, often resulting in self-digestion, edema, hemorrhage, and even necrosis of pancreatic tissue. When AP progresses to severe acute pancreatitis (SAP), it often causes multi-organ damage, leading to a high mortality rate. However, the molecular mechanisms underlying SAP-mediated organ damage remain unclear. This study aims to systematically mine SAP data from public databases and combine experimental validation to identify key molecules involved in multi-organ damage caused by SAP. Retrieve transcriptomic data of mice pancreatic tissue for AP, lung and liver tissue for SAP, and corresponding normal tissue from the Gene Expression Omnibus (GEO) database. Conduct gene differential analysis using Limma and DEseq2 methods. Perform enrichment analysis using the clusterProfiler package in R software. Score immune cells and immune status in various organs using single-sample gene set enrichment analysis (ssGSEA). Evaluate mRNA expression levels of core genes using reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. Validate serum amylase, TNF-α, IL-1β, and IL-6 levels in peripheral blood using enzyme-linked immunosorbent assay (ELISA), and detect the formation of neutrophil extracellular traps (NETs) in mice pancreatic, liver, and lung tissues using immunofluorescence. Differential analysis reveals that 46 genes exhibit expression dysregulation in mice pancreatic tissue for AP, liver and lung tissue for SAP, as well as peripheral blood in humans. Functional enrichment analysis indicates that these genes are primarily associated with neutrophil-related biological processes. ROC curve analysis indicates that 12 neutrophil-related genes have diagnostic potential for SAP. Immune infiltration analysis reveals high neutrophil infiltration in various organs affected by SAP. Single-cell sequencing analysis shows that these genes are predominantly expressed in neutrophils and macrophages. FPR1, ITGAM, and C5AR1 are identified as key genes involved in the formation of NETs and activation of neutrophils. qPCR and IHC results demonstrate upregulation of FPR1, ITGAM, and C5AR1 expression in pancreatic, liver, and lung tissues of mice with SAP. Immunofluorescence staining shows increased levels of neutrophils and NETs in SAP mice. Inhibition of NETs formation can alleviate the severity of SAP as well as the levels of inflammation in the liver and lung tissues. This study identified key genes involved in the formation of NETs, namely FPR1, ITGAM, and C5AR1, which are upregulated during multi-organ damage in SAP. Inhibition of NETs release effectively reduces the systemic inflammatory response and liver-lung damage in SAP. This research provides new therapeutic targets for the multi-organ damage associated with SAP.
Collapse
Affiliation(s)
- Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziang Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heming Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingmei Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
29
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2025; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
30
|
Rodrigues KB, Weng Z, Graham ZA, Lavin K, McAdam J, Tuggle SC, Peoples B, Seay R, Yang S, Bamman MM, Broderick TJ, Montgomery SB. Exercise intensity and training alter the innate immune cell type and chromosomal origins of circulating cell-free DNA in humans. Proc Natl Acad Sci U S A 2025; 122:e2406954122. [PMID: 39805013 PMCID: PMC11761974 DOI: 10.1073/pnas.2406954122] [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: 05/05/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Exercising regularly promotes health, but these benefits are complicated by acute inflammation induced by exercise. A potential source of inflammation is cell-free DNA (cfDNA), yet the cellular origins, molecular causes, and immune system interactions of exercise-induced cfDNA are unclear. To study these, 10 healthy individuals were randomized to a 12-wk exercise program of either high-intensity tactical training (HITT) or traditional moderate-intensity training (TRAD). Blood plasma was collected pre- and postexercise at weeks 0 and 12 and after 4 wk of detraining upon program completion. Whole-genome enzymatic methylation sequencing (EM-seq) with cell-type proportion deconvolution was applied to cfDNA obtained from the 50 plasma samples and paired to concentration measurements for 90 circulating cytokines. Acute exercise increased the release of cfDNA from neutrophils, dendritic cells (DCs), and macrophages proportional to exercise intensity. Exercise training reduced cfDNA released in HITT participants but not TRAD and from DCs and macrophages but not neutrophils. For most participants, training lowered mitochondrial cfDNA at rest, even after detraining. Using a sequencing analysis approach we developed, we concluded that rapid ETosis, a process of cell death where cells release DNA extracellular traps, was the likely source of cfDNA, demonstrated by enrichment of nuclear DNA. Further, several cytokines were induced by acute exercise, such as IL-6, IL-10, and IL-16, and training attenuated the induction of only IL-6 and IL-17F. Cytokine levels were not associated with cfDNA induction, suggesting that these cytokines are not the main cause of exercise-induced cfDNA. Overall, exercise intensity and training modulated cfDNA release and cytokine responses, contributing to the anti-inflammatory effects of regular exercise.
Collapse
Affiliation(s)
- Kameron B. Rodrigues
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Ziming Weng
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Zachary A. Graham
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | - Kaleen Lavin
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | - Jeremy McAdam
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | - S. Craig Tuggle
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | - Brandon Peoples
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL35294
| | - Regina Seay
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL35294
| | - Sufen Yang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL35294
| | - Marcas M. Bamman
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | - Timothy J. Broderick
- Healthspan, Resilience and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, FL32502
| | | |
Collapse
|
31
|
Feješ A, Šebeková K, Borbélyová V. Pathophysiological Role of Neutrophil Extracellular Traps in Diet-Induced Obesity and Metabolic Syndrome in Animal Models. Nutrients 2025; 17:241. [PMID: 39861371 PMCID: PMC11768048 DOI: 10.3390/nu17020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The global pandemic of obesity poses a serious health, social, and economic burden. Patients living with obesity are at an increased risk of developing noncommunicable diseases or to die prematurely. Obesity is a state of chronic low-grade inflammation. Neutrophils are first to be recruited to sites of inflammation, where they contribute to host defense via phagocytosis, degranulation, and extrusion of neutrophil extracellular traps (NETs). NETs are web-like DNA structures of nuclear or mitochondrial DNA associated with cytosolic antimicrobial proteins. The primary function of NETosis is preventing the dissemination of pathogens. However, neutrophils may occasionally misidentify host molecules as danger-associated molecular patterns, triggering NET formation. This can lead to further recruitment of neutrophils, resulting in propagation and a vicious cycle of persistent systemic inflammation. This scenario may occur when neutrophils infiltrate expanded obese adipose tissue. Thus, NETosis is implicated in the pathophysiology of autoimmune and metabolic disorders, including obesity. This review explores the role of NETosis in obesity and two obesity-associated conditions-hypertension and liver steatosis. With the rising prevalence of obesity driving research into its pathophysiology, particularly through diet-induced obesity models in rodents, we discuss insights gained from both human and animal studies. Additionally, we highlight the potential offered by rodent models and the opportunities presented by genetically modified mouse strains for advancing our understanding of obesity-related inflammation.
Collapse
Affiliation(s)
| | - Katarína Šebeková
- Institute of Molecular Biomedicine, Medical Faculty, Comenius University, 83303 Bratislava, Slovakia; (A.F.); (V.B.)
| | | |
Collapse
|
32
|
Jarzebska N, Rodionov RN, Voit-Bak K, Straube R, Mücke A, Tselmin S, Rettig R, Julius U, Siow R, Gräßler J, Passauer J, Kok Y, Mavberg P, Weiss N, Bornstein SR, Aswani A. Neutrophil Extracellular Traps (NETs) as a Potential Target for Anti-Aging: Role of Therapeutic Apheresis. Horm Metab Res 2025. [PMID: 39788160 DOI: 10.1055/a-2444-3422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Neutrophil extracellular traps (NETs) are large structures composed of chromatin, histones and granule-derived proteins released extracellularly by neutrophils. They are generally considered to be a part of the antimicrobial defense strategy, preventing the dissemination of pathogens. However, overproduction of NETs or their ineffective clearance can drive various pathologies, many of which are associated with advanced age and involve uncontrolled inflammation, oxidative, cardiovascular and neurodegenerative stress as underlying mechanisms. Targeting NETs in the elderly as an anti-aging therapy seems to be a very attractive therapeutic approach. Therapeutic apheresis with a specific filter to remove NETs could be a promising strategy worth considering.
Collapse
Affiliation(s)
- Natalia Jarzebska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Karin Voit-Bak
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Richard Straube
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Anna Mücke
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Sergey Tselmin
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ronny Rettig
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ulrich Julius
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Richard Siow
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Ageing Research at King's (ARK), King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Jürgen Gräßler
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jens Passauer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | | | - Philip Mavberg
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Norbert Weiss
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Aswani
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom of Great Britain and Northern Ireland
- Santersus AG, Zurich, Switzerland
| |
Collapse
|
33
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
34
|
Becker RC, Tantry US, Khan M, Gurbel PA. The COVID-19 thrombus: distinguishing pathological, mechanistic, and phenotypic features and management. J Thromb Thrombolysis 2025; 58:15-49. [PMID: 39179952 PMCID: PMC11762605 DOI: 10.1007/s11239-024-03028-4] [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] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
A heightened risk for thrombosis is a hallmark of COVID-19. Expansive clinical experience and medical literature have characterized small (micro) and large (macro) vessel involvement of the venous and arterial circulatory systems. Most events occur in patients with serious or critical illness in the hyperacute (first 1-2 weeks) or acute phases (2-4 weeks) of SARS-CoV-2 infection. However, thrombosis involving the venous, arterial, and microcirculatory systems has been reported in the subacute (4-8 weeks), convalescent (> 8-12 weeks) and chronic phases (> 12 weeks) among patients with mild-to-moderate illness. The purpose of the current focused review is to highlight the distinguishing clinical features, pathological components, and potential mechanisms of venous, arterial, and microvascular thrombosis in patients with COVID-19. The overarching objective is to better understand the proclivity for thrombosis, laying a solid foundation for screening and surveillance modalities, preventive strategies, and optimal patient management.
Collapse
Affiliation(s)
- Richard C Becker
- Cardiovascular Center, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| | - Muhammad Khan
- Division of General Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| |
Collapse
|
35
|
Cao X, Lan Q, Xu H, Liu W, Cheng H, Hu X, He J, Yang Q, Lai W, Chu Z. Granulocyte-like myeloid-derived suppressor cells: The culprits of neutrophil extracellular traps formation in the pre-metastatic niche. Int Immunopharmacol 2024; 143:113500. [PMID: 39510029 DOI: 10.1016/j.intimp.2024.113500] [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: 08/10/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Neutrophil extracellular traps (NETs) have been shown to exhibit chemotactic effects on circulating tumor cells at metastatic sites, promoting the progression of colorectal cancer liver metastasis (CRLM). However, the origin and factors contributing to the formation of NETs (NETosis) in the pre-metastatic niche (PMN) of target organs remain unclear. In this study, we investigated the relationship between phosphatase of regenerating liver-3 (PRL-3), myeloid-derived suppressor cells (MDSCs), neutrophils, and NETs through a retrospective clinical cohort study and a mouse model of CRLM. Our clinical findings revealed associations between PRL-3 expression and the infiltration of neutrophils and MDSCs in CRLM patients. Moreover, NETosis emerged as a robust indicator of poor prognosis for overall survival in these patients. In CRLM models, PRL-3 overexpression enhanced both primary tumor growth and liver metastasis. We found that the first week after tumor cell implantation appeared to be a crucial period for PMN formation, with notable infiltration of neutrophils, MDSCs, and NETosis during this time. Notably, co-culturing neutrophils with MDSCs induced NETosis in vitro, particularly with granulocyte-like MDSCs (Gr-MDSCs), the predominant subtype of infiltrating MDSCs. In summary, our findings elucidate the likely origin of NETs in the PMN during CRLM development and underscore the significant influence of PRL-3. These findings may offer potential immunotherapeutic targets for patients at risk of developing CRLM, warranting further investigation in clinical settings.
Collapse
Affiliation(s)
- Xintong Cao
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Qiusheng Lan
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Heyang Xu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Wentao Liu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Huaxi Cheng
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Xinwen Hu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Jiehua He
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, Guangdong Province, China
| | - Qiong Yang
- Department of Medical Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| | - Wei Lai
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| | - Zhonghua Chu
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China.
| |
Collapse
|
36
|
Altamura S, Lombardi F, Palumbo P, Cinque B, Ferri C, Del Pinto R, Pietropaoli D. The Evolving Role of Neutrophils and Neutrophil Extracellular Traps (NETs) in Obesity and Related Diseases: Recent Insights and Advances. Int J Mol Sci 2024; 25:13633. [PMID: 39769394 PMCID: PMC11727698 DOI: 10.3390/ijms252413633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
Obesity is a chronic, multifactorial disease characterized by persistent low-grade tissue and systemic inflammation. Fat accumulation in adipose tissue (AT) leads to stress and dysfunctional adipocytes, along with the infiltration of immune cells, which initiates and sustains inflammation. Neutrophils are the first immune cells to infiltrate AT during high-fat diet (HFD)-induced obesity. Emerging evidence suggests that the formation and release of neutrophil extracellular traps (NETs) play a significant role in the progression of obesity and related diseases. Additionally, obesity is associated with an imbalance in gut microbiota and increased intestinal barrier permeability, resulting in the translocation of live bacteria, bacterial deoxyribonucleic acid (DNA), lipopolysaccharides (LPS), and pro-inflammatory cytokines into the bloodstream and AT, thereby contributing to metabolic inflammation. Recent research has also shown that short-chain fatty acids (SCFAs), produced by gut microbiota, can influence various functions of neutrophils, including their activation, migration, and the generation of inflammatory mediators. This review comprehensively summarizes recent advancements in understanding the role of neutrophils and NET formation in the pathophysiology of obesity and related disorders while also focusing on updated potential therapeutic approaches targeting NETs based on studies conducted in humans and animal models.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| |
Collapse
|
37
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
38
|
Sun R, Chu J, Li P. Inflammasomes and idiopathic inflammatory myopathies. Front Immunol 2024; 15:1449969. [PMID: 39723212 PMCID: PMC11668653 DOI: 10.3389/fimmu.2024.1449969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIM) are a group of systemic autoimmune diseases characterized by muscle weakness and elevated serum creatine kinase levels. Recent research has highlighted the role of the innate immune system, particularly inflammasomes, in the pathogenesis of IIM. This review focuses on the role of inflammasomes, specifically NLRP3 and AIM2, and their associated proteins in the development of IIM. We discuss the molecular mechanisms of pyroptosis, a programmed cell death pathway that triggers inflammation, and its association with IIM. The NLRP3 inflammasome, in particular, has been implicated in muscle fiber necrosis and the subsequent release of damage-associated molecular patterns (DAMPs), leading to inflammation. We also explore the potential therapeutic implications of targeting the NLRP3 inflammasome with inhibitors such as glyburide and MCC950, which have shown promise in reducing inflammation and improving muscle function in preclinical models. Additionally, we discuss the role of caspases, particularly caspase-1, in the canonical pyroptotic pathway associated with IIM. The understanding of these mechanisms offers new avenues for therapeutic intervention and a better comprehension of IIM pathophysiology.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jiyan Chu
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
- Graduate School, Dalian Medical University, Dalian, Liaoning, China
| | - Ping Li
- Department of Rheumatology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
39
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
40
|
Yipeng Z, Chao C, Ranran L, Tingting P, Hongping Q. Metabolism: a potential regulator of neutrophil fate. Front Immunol 2024; 15:1500676. [PMID: 39697327 PMCID: PMC11652355 DOI: 10.3389/fimmu.2024.1500676] [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: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are essential components of the innate immune system that defend against the invading pathogens, such as bacteria, viruses, and fungi, as well as having regulatory roles in various conditions, including tissue repair, cancer immunity, and inflammation modulation. The function of neutrophils is strongly related to their mode of cell death, as different types of cell death involve various cellular and molecular alterations. Apoptosis, a non-inflammatory and programmed type of cell death, is the most common in neutrophils, while other modes of cell death, including NETOsis, necrosis, necroptosis, autophagy, pyroptosis, and ferroptosis, have specific roles in neutrophil function regulation. Immunometabolism refers to energy and substance metabolism in immune cells, and profoundly influences immune cell fate and immune system function. Intercellular and intracellular signal transduction modulate neutrophil metabolism, which can, in turn, alter their activities by influencing various cell signaling pathways. In this review, we compile an extensive body of evidence demonstrating the role of neutrophil metabolism in their various forms of cell death. The review highlights the intricate metabolic characteristics of neutrophils and their interplay with various types of cell death.
Collapse
Affiliation(s)
| | | | | | - Pan Tingting
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| | - Qu Hongping
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Li J, Zhao H, Yang J, Wang M, Cao Z, Wang Y, Gu Z. The role and mechanism of extracellular traps in chronic rhinosinusitis. Biomed Pharmacother 2024; 181:117655. [PMID: 39486368 DOI: 10.1016/j.biopha.2024.117655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/24/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Chronic rhinosinusitis (CRS) is a common inflammatory disease of the nose that affects millions of individuals worldwide. Recent research has introduced the concept of an immunologic endotype based on the pathological characteristics of CRS and the types of inflammatory cell infiltration. This endotype concept is conducive to understanding CRS pathology and guiding further targeted therapy. Eosinophils and neutrophils infiltrate different proportions in different CRS endotypes and release extracellular traps (ETs) as a response to the extracellular immune response. The mechanisms of formation and biological roles of ETs are complex. ETs can trap extracellular microorganisms and limit the range of inflammation to some extent; however, excessive and long-term ETs may be related to disease severity. This review summarises and explores the mechanism of ETs and the advances in CRS research and proposes new insights into the interaction between ETs and programmed cell death (including autophagy, pyroptosis, and necroptosis) in CRS, providing new ideas for the targeted therapy of CRS.
Collapse
Affiliation(s)
- Jiani Li
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - He Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Meng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Zhiwei Cao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Yunxiu Wang
- Department of Clinical Trial Ward, Clinical Trial and Conversion Center, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Zhaowei Gu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
42
|
Prendecki M, Gurung A, Pisacano N, Pusey CD. The role of neutrophils in ANCA-associated vasculitis. Immunol Lett 2024; 270:106933. [PMID: 39362307 DOI: 10.1016/j.imlet.2024.106933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) is a group of rare systemic autoimmune diseases characterised by necrotising inflammation of small blood vessels and usually associated with circulating ANCA. The pathophysiology of AAV is complex, involving many aspects of the innate and adaptive immune system. Neutrophils are central to the pathogenesis of AAV as they are both the target of the autoantibody and effector cells mediating vascular injury. We describe mechanisms for ANCA induced activation of neutrophils, the pathogenic mechanisms by which this leads to endothelial cell injury, and how neutrophil crosstalk modulates other aspects of the immune system in AAV.
Collapse
Affiliation(s)
- Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom.
| | - Angila Gurung
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Noelle Pisacano
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
43
|
Balog BM, Niemi JP, Disabato T, Hashim F, Zigmond RE. CXCR2 mediated trafficking of neutrophils and neutrophil extracellular traps are required for myelin clearance after a peripheral nerve injury. Exp Neurol 2024; 382:114985. [PMID: 39368532 PMCID: PMC11526632 DOI: 10.1016/j.expneurol.2024.114985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Neutrophils are a vital part of the innate immune system. Many of their functions eliminate bacteria & viruses, like neutrophil extracellular traps (NETs), which trap bacteria, enhancing macrophage phagocytosis. It was surprising when it was demonstrated that neutrophils are a part of Wallerian degeneration, a process that is essential for nerve regeneration after a nerve injury. It is not known what signals attract neutrophils into the nerve and how they aid Wallerian degeneration. Neutrophils accumulate in the distal nerve within one day after an injury and are found in the nerve from one to three days. We demonstrate that CXCR2 mediates the trafficking of neutrophils into the distal nerve, and without CXCR2 Wallerian degeneration, as indicated by luxol fast blue staining, was reduced seven days after a sciatic nerve crush or transection injury. NETs were detected in the distal nerve after a sciatic nerve transection. NET formation has been shown to require protein arginine deiminase 4 (PAD4), which citrullinates histone 3. Inhibiting PAD4 reduced NET formation significantly in the distal nerve at two days and myelin clearance at seven days indicating that NETs aid myelin clearance. These results demonstrate another function for NETs other than clearing pathogens. Neutrophils have been detected after injuries to the central nervous system and diseases in humans and animal models. Our results demonstrate neutrophils aid myelin clearance, suggesting a role for their presence in central nervous system injuries and diseases.
Collapse
Affiliation(s)
- Brian M Balog
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Thomas Disabato
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Faye Hashim
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
44
|
Wu Y, Shen J. Unraveling the intricacies of neutrophil extracellular traps in inflammatory bowel disease: Pathways, biomarkers, and promising therapies. Cytokine Growth Factor Rev 2024; 80:156-167. [PMID: 39438227 DOI: 10.1016/j.cytogfr.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
The development of inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, involves various factors and is characterized by persistent inflammation of the mucosal lining. However, the role of neutrophils in this process remains controversial. Neutrophil extracellular traps (NETs), which consist of chromatin, antimicrobial proteins, and oxidative enzymes, are released by neutrophils to trap pathogens. They are also involved in various immune-mediated and vascular diseases. NETs act as a vital defense mechanisms at the gut-mucosal interface and are frequently exposed to bacterial, viral, and fungal threats. However, they can also contribute to inflammation and worsen imbalances in the gut bacteria. Recent studies have suggested that NETs have a significant impact on IBD development. Previous studies have shown increased levels of NETs in tissue and blood samples from patients with IBD, as well as in experimental colitis mouse models. Therefore, this review discusses how NETs are formed and their role in the pathophysiology of IBD. It discusses how NETs may lead to tissue damage and contribute to IBD-associated complications. Moreover, non-invasive biomarkers are needed to replace invasive procedures such as endoscopy to better evaluate the disease status. Given the crucial role of NETs in IBD progression, this review focuses on potential NET biomarkers that can help predict the evolution of IBD. Furthermore, this review identifies potential therapeutic targets for regulating NET production, which could expand the range of available treatment options for IBD.
Collapse
Affiliation(s)
- Yilin Wu
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai 200127, China; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China; Shanghai Institute of Digestive Disease, No.160 PuJian Road, China.
| |
Collapse
|
45
|
Wang Q, Wei J, He J, Ming S, Li X, Huang X, Hong Z, Wu Y. HSP70 contributes to pathogenesis of fulminant hepatitis induced by coronavirus. Int Immunopharmacol 2024; 141:112963. [PMID: 39159560 DOI: 10.1016/j.intimp.2024.112963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/07/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
Fulminant viral hepatitis (FH) represents a significant clinical challenge, with its pathogenesis not yet fully elucidated. Heat shock protein (HSP)70, a molecular chaperone protein with a broad range of cytoprotective functions, is upregulated in response to stress. However, the role of HSP70 in FH remains to be investigated. Notably, HSP70 expression is upregulated in the livers of coronavirus-infected mice and patients. Therefore, we investigated the mechanistic role of HSP70 in coronavirus-associated FH pathogenesis. FH was induced in HSP70-deficient (HSP70 KO) mice or in WT mice treated with the HSP70 inhibitor VER155008 when infected with the mouse hepatitis virus strain A59 (MHV-A59). MHV-A59-infected HSP70 KO mice exhibited significantly reduced liver damage and mortality. This effect was attributed to decreased infiltration of monocyte-macrophages and neutrophils in the liver of HSP70 KO mice, resulting in lower levels of inflammatory cytokines such as IL-1β, TNFα, and IL-6, and a reduced viral load. Moreover, treatment with the HSP70 inhibitor VER155008 protected mice from MHV-A59-induced liver damage and FH mortality. In summary, HSP70 promotes coronavirus-induced FH pathogenesis by enhancing the infiltration of monocyte-macrophages and neutrophils and promoting the secretion of inflammatory cytokines. Therefore, HSP70 is a potential therapeutic target in viral FH intervention.
Collapse
Affiliation(s)
- Qiaohua Wang
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jiayou Wei
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Siqi Ming
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, Guangdong Province 519015, China
| | - Xingyu Li
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xi Huang
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhongsi Hong
- Center of Infectious Disease, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| | - Yongjian Wu
- Center for Infection and Immunity, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| |
Collapse
|
46
|
Watanabe Y, Obama T, Makiyama T, Itabe H. Oxysterols Suppress Release of DNA from Granulocytes into Extracellular Space After Stimulation with Phorbol Myristate Acetate. Biomedicines 2024; 12:2535. [PMID: 39595101 PMCID: PMC11592087 DOI: 10.3390/biomedicines12112535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Neutrophils eject their DNA strings and cellular proteins into the extracellular space upon treatment with various stimulants. In the present study, we examined the effects of four major oxidized cholesterol metabolites on DNA release from granulocytes. METHODS AND RESULTS When oxysterols were added to HL-60-derived granulocytes stimulated with phorbol 12-myristate 13-acetate (PMA), they suppressed the release of DNA and myeloperoxidase from the cells. Among the four oxysterols tested, 7-ketocholesterol was the most effective. Addition of the same concentration of 7-ketocholesterol did not induce any cytotoxic effects, as evaluated based on the release of lactate dehydrogenase and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazoliumbromide (MTT) assays. DNA release from human peripheral blood neutrophils after PMA stimulation was also suppressed by 7-ketocholesterol. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to quantify sterol content in the cells. The addition of oxysterols increased the cellular content of the corresponding compounds by more than 10-fold compared to those at baseline. Treatment of HL-60-derived granulocytes with methyl-β-cyclodextrin that removes sterol compounds from the membranes increased DNA release from the cells in a dose-dependent manner. CONCLUSIONS These results suggest that oxysterols have suppressive effects on DNA release from granulocytes stimulated with PMA.
Collapse
Affiliation(s)
| | | | | | - Hiroyuki Itabe
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; (Y.W.); (T.O.); (T.M.)
| |
Collapse
|
47
|
Chen H, Zhou Y, Tang Y, Lan J, Lin C, Chen Q, Kuang H. Neutrophil extracellular traps in tumor progression of gynecologic cancers. Front Immunol 2024; 15:1421889. [PMID: 39555072 PMCID: PMC11563837 DOI: 10.3389/fimmu.2024.1421889] [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: 04/23/2024] [Accepted: 09/30/2024] [Indexed: 11/19/2024] Open
Abstract
This article delves into the intricate interplay between tumors, particularly gynecologic malignancies, and neutrophil extracellular traps (NETs). The relationship between tumors, specifically gynecologic malignancies, and NETs is a multifaceted and pivotal area of study. Neutrophils, pivotal components of the immune system, are tasked with combating foreign invaders. NETs, intricate structures released by neutrophils, play a vital role in combating systemic infections but also play a role in non-infectious conditions such as inflammation, autoimmune diseases, and cancer. Cancer cells have the ability to attract neutrophils, creating tumor-associated neutrophils, which then stimulate the release of NETs into the tumor microenvironment. The impact of NETs within the tumor microenvironment is profound and intricate. They play a significant role in influencing cancer development and metastasis, as well as modulating tumor immune responses. Through the release of proteases and pro-inflammatory cytokines, NETs directly alter the behavior of tumor cells, increasing invasiveness and metastatic potential. Additionally, NETs can trigger epithelial-mesenchymal transition in tumor cells, a process associated with increased invasion and metastasis. The interaction between tumors and NETs is particularly critical in gynecologic malignancies such as ovarian, cervical, and endometrial cancer. Understanding the mechanisms through which NETs operate in these tumors can offer valuable insights for the development of targeted therapeutic interventions. Researchers are actively working towards harnessing this interaction to impede tumor progression and metastasis, opening up new avenues for future treatment modalities. As our understanding of the interplay between tumors and NETs deepens, it is anticipated that novel treatment strategies will emerge, potentially leading to improved outcomes for patients with gynecologic malignancies. This article provides a comprehensive overview of the latest research findings on the interaction between NETs and cancer, particularly in gynecologic tumors, serving as a valuable resource for future exploration in this field.
Collapse
Affiliation(s)
- Hong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yaling Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianfa Lan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chao Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qionghua Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongying Kuang
- The Second Department of Gynecology, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
48
|
Pituk D, Balogh L, Horváth E, Hegyi Z, Baráth B, Bogáti R, Szűcs P, Papp Z, Katona É, Bereczky Z. Localization of Hemostasis Elements in Aspirated Coronary Thrombi at Different Stages of Evolution. Int J Mol Sci 2024; 25:11746. [PMID: 39519297 PMCID: PMC11547099 DOI: 10.3390/ijms252111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The structure of aspirated coronary thrombus in ST-segment elevation myocardial infarction (STEMI) is still being studied. Our aims were to characterize coronary thrombi of different ages, focusing on the appearance of activated protein C (APC/PC) and its relation to the elements of neutrophil extracellular traps (NETs), and the factors closely related to fibrin as factor XIII (FXIII) and α2 plasmin inhibitor (α2-PI). The thrombi of n = 24 male patients with atherosclerotic coronary plaque rupture related to native coronary artery occlusion were selected for histopathology analysis. Thrombus age was distinguished as fresh, lytic, and organized, and then analyzed by immunofluorescent staining and confocal microscopy. FXIII was present at a high level and showed a high degree of co-localization with fibrin in all stages of thrombus evolution. The amount of α2-PI was low in the fresh thrombi, which increased significantly to the lytic phase. It was evenly distributed and consistently associated with fibrin. APC/PC appeared in the fresh thrombus and remained constant during its evolution. The presence of NET marker and CD66b was most dominant in the lytic phase. APC/PC co-localization with the elements of NET formation shows its role in NET degradation. These observations suggest the importance of searching for further targeted therapeutic strategies in STEMI patients.
Collapse
Affiliation(s)
- Dóra Pituk
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.P.); (B.B.); (R.B.); (É.K.)
- Kálmán Laki Doctoral School, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Balogh
- Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (L.B.); (Z.P.)
| | - Emőke Horváth
- Department of Pathology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Zoltán Hegyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.H.); (P.S.)
| | - Barbara Baráth
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.P.); (B.B.); (R.B.); (É.K.)
| | - Réka Bogáti
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.P.); (B.B.); (R.B.); (É.K.)
| | - Péter Szűcs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.H.); (P.S.)
| | - Zoltán Papp
- Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (L.B.); (Z.P.)
| | - Éva Katona
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.P.); (B.B.); (R.B.); (É.K.)
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.P.); (B.B.); (R.B.); (É.K.)
| |
Collapse
|
49
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024; 38:5067-5087. [PMID: 39105461 DOI: 10.1002/ptr.8311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
50
|
Wu G, Pan B, Shi H, Yi Y, Zheng X, Ma H, Zhao M, Zhang Z, Cheng L, Huang Y, Guo W. Neutrophils' dual role in cancer: from tumor progression to immunotherapeutic potential. Int Immunopharmacol 2024; 140:112788. [PMID: 39083923 DOI: 10.1016/j.intimp.2024.112788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
The tumor microenvironment (TME) is intricately associated with cancer progression, characterized by dynamic interactions among various cellular and molecular components that significantly impact the carcinogenic process. Notably, neutrophils play a crucial dual role in regulating this complex environment. These cells oscillate between promoting and inhibiting tumor activity, responding to a multitude of cytokines, chemokines, and tumor-derived factors. This response modulates immune reactions and affects the proliferation, metastasis, and angiogenesis of cancer cells. A significant aspect of their influence is their interaction with the endoplasmic reticulum (ER) stress responses in cancer cells, markedly altering tumor immunodynamics by modulating the phenotypic plasticity and functionality of neutrophils. Furthermore, neutrophil extracellular traps (NETs) exert a pivotal influence in the progression of malignancies by enhancing inflammation, metastasis, immune suppression, and thrombosis, thereby exacerbating the disease. In the realm of immunotherapy, checkpoint inhibitors targeting PD-L1/PD-1 and CTLA-4 among others have underscored the significant role of neutrophils in enhancing therapeutic responses. Recent research has highlighted the potential of using neutrophils for targeted drug delivery through nanoparticle systems, which precisely control drug release and significantly enhance antitumor efficacy. This review thoroughly examines the diverse functions of neutrophils in cancer treatment, emphasizing their potential in regulating immune therapy responses and as drug delivery carriers, offering innovative perspectives and profound implications for the development of targeted diagnostic and therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaobin Zheng
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, China
| | - Lin Cheng
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI), Galway, Ireland.
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weigang Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|