1
|
Lu Z, Zhu L, Yi C, Su B, Wang R. C5a/C5aR regulates Th1/Th2 imbalance in sepsis-associated lung injury by promoting neutrophil activation to increase PAD4 expression. Ann Med 2025; 57:2447406. [PMID: 39831526 PMCID: PMC11749016 DOI: 10.1080/07853890.2024.2447406] [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: 10/25/2023] [Revised: 08/09/2024] [Accepted: 09/13/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Multi-organ failure frequently complicates sepsis, with lungs being the primary target. T helper (Th) cell activation and phenotypic imbalance among them contribute significantly to sepsis-associated lung injury. Additionally, the complement system could regulate the polarized phenotype of T lymphocytes. Therefore, this study investigated the effect of C5a/C5a receptor (C5aR)/Peptidylarginine deiminase 4 (PAD4) on the Th1/Th2 ratio in sepsis-induced lung injury. METHODS ELISA was used to detect the expression of PAD4, HBP, MPO, IL-1β, IL-10, IL-6, IL-4, syndecan-1, endocan and H3Cit. An LPS-induced septic lung injury mouse model was constructed, with HE and PAS stains evaluating lung damage. BCA kit quantified BALF total protein, Western blot examined C5aR, syndecan-1, endocan, PAD4 levels, while TUNEL and flow cytometry assessed tissue cellular apoptosis. Furthermore, flow cytometry was used to detect the +Th1 and Th2 cells proportion in peripheral blood, and CCK-8 was used to detect BEAS-2B activity. RESULTS The results indicated that PAD4 and inflammatory factors were increased in lesion samples compared with controls. In sepsis-induced lung injury mice, addition of GSK484, a PAD4 inhibitor, effectively alleviated sepsis-induced lung edema and inflammatory responses. GSK484 was found to inhibit C5a/C5aR expression and suppress apoptosis and lung injury. Furthermore, GSK484 markedly inhibited Th1 cell phenotypes in vitro. Additionally, GSK484 intervention on Th1 cell phenotype further affected lung epithelial cell injury. CONCLUSION In summary, we revealed the mechanism of C5a/C5aR-induced PAD4 upregulation via neutrophil activation in sepsis-associated lung injury, causing a Th1/Th2 imbalance and lung injury, providing a novel approach for sepsis-associated lung injuries treatment.
Collapse
Affiliation(s)
- Zhenbing Lu
- Department of Emergency, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Zhu
- Department of Emergency, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Changlin Yi
- Department of Clinical Laboratory, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bi Su
- Department of Emergency, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Renying Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Chen SH, Chen CH, Lin HC, Yeh SA, Hwang TL, Chen PJ. Drug repurposing of cyclin-dependent kinase inhibitors for neutrophilic acute respiratory distress syndrome and psoriasis. J Adv Res 2025; 72:485-500. [PMID: 39089617 DOI: 10.1016/j.jare.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Neutrophilic inflammation, characterized by dysregulated neutrophil activation, triggers a variety of inflammatory responses such as chemotactic infiltration, oxidative bursts, degranulation, neutrophil extracellular traps (NETs) formation, and delayed turnover. This type of inflammation is pivotal in the pathogenesis of acute respiratory distress syndrome (ARDS) and psoriasis. Despite current treatments, managing neutrophil-associated inflammatory symptoms remains a significant challenge. AIM OF REVIEW This review emphasizes the role of cyclin-dependent kinases (CDKs) in neutrophil activation and inflammation. It aims to highlight the therapeutic potential of repurposing CDK inhibitors to manage neutrophilic inflammation, particularly in ARDS and psoriasis. Additionally, it discusses the necessary precautions for the clinical application of these inhibitors due to potential off-target effects and the need for dose optimization. KEY SCIENTIFIC CONCEPTS OF REVIEW CDKs regulate key neutrophilic functions, including chemotactic responses, degranulation, NET formation, and apoptosis. Repurposing CDK inhibitors, originally developed for cancer treatment, shows promise in controlling neutrophilic inflammation. Clinical anticancer drugs, palbociclib and ribociclib, have demonstrated efficacy in treating neutrophilic ARDS and psoriasis by targeting off-label pathways, phosphoinositide 3-kinase (PI3K) and phosphodiesterase 4 (PDE4), respectively. While CDK inhibitors offer promising therapeutic benefits, their clinical repurposing requires careful consideration of off-target effects and dose optimization. Further exploration and clinical trials are necessary to ensure their safety and efficacy in treating inflammatory conditions.
Collapse
Affiliation(s)
- Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan.
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Hsin-Chieh Lin
- Department of Chinese Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824410, Taiwan; School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Shyh-An Yeh
- Medical Physics and Informatics Laboratory of Electronic Engineering and Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan; Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung 824410, Taiwan; Department of Radiation Oncology, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan.
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan; Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan.
| |
Collapse
|
3
|
Qu G, Ribeiro HA, Solomon AL, Sordo Vieira L, Goddard Y, Diodati NG, Lazarte AV, Wheeler M, Laubenbacher R, Mehrad B. The heme scavenger hemopexin protects against lung injury during aspergillosis by mitigating release of neutrophil extracellular traps. JCI Insight 2025; 10:e189151. [PMID: 40232861 DOI: 10.1172/jci.insight.189151] [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: 11/12/2024] [Accepted: 04/09/2025] [Indexed: 04/17/2025] Open
Abstract
Invasive aspergillosis is characterized by lung hemorrhage and release of extracellular heme, which promotes fungal growth. Heme can also mediate tissue injury directly, and both fungal growth and lung injury may induce hemorrhage. To assimilate these interdependent processes, we hypothesized that, during aspergillosis, heme mediates direct lung injury independent of fungal growth, leading to worse infection outcomes, and the scavenger protein hemopexin mitigates these effects. Mice with neutropenic aspergillosis developed a time-dependent increase in lung extracellular heme and a corresponding hemopexin induction. Hemopexin deficiency resulted in markedly increased lung injury, fungal growth, and lung hemorrhage. Using a computational model of the interactions of Aspergillus, heme, and the host, we predicted a critical role for heme-mediated generation of neutrophil extracellular traps (NETs) in this infection. We tested this prediction using a fungal strain unable to grow at body temperature and found that extracellular heme and fungal exposure synergized to induce lung injury by promoting NET release, and disruption of NET was sufficient to attenuate lung injury and fungal burden. These data implicate heme-mediated NETosis in both lung injury and fungal growth during aspergillosis, resulting in a detrimental positive feedback cycle that can be interrupted by scavenging heme or disrupting NETs.
Collapse
|
4
|
Cleary SJ, Qiu L, Seo Y, Baluk P, Liu D, Serwas NK, Taylor CA, Zhang D, Cyster JG, McDonald DM, Krummel MF, Looney MR. Intravital imaging of pulmonary lymphatics in inflammation and metastatic cancer. J Exp Med 2025; 222:e20241359. [PMID: 39969509 PMCID: PMC11837973 DOI: 10.1084/jem.20241359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/11/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025] Open
Abstract
Intravital microscopy has enabled the study of immune dynamics in the pulmonary microvasculature, but many key events remain unseen because they occur in deeper lung regions. We therefore developed a technique for stabilized intravital imaging of bronchovascular cuffs and collecting lymphatics surrounding pulmonary veins in mice. Intravital imaging of pulmonary lymphatics revealed ventilation dependence of steady-state lung lymph flow and ventilation-independent lymph flow during inflammation. We imaged the rapid exodus of migratory dendritic cells through lung lymphatics following inflammation and measured effects of pharmacologic and genetic interventions targeting chemokine signaling. Intravital imaging also captured lymphatic immune surveillance of lung-metastatic cancers and lymphatic metastasis of cancer cells. To our knowledge, this is the first imaging of lymph flow and leukocyte migration through intact pulmonary lymphatics. This approach will enable studies of protective and maladaptive processes unfolding within the lungs and in other previously inaccessible locations.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Dan Liu
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
- Westlake Laboratory of Life Sciences and Biomedicine, School of Medicine, Westlake University, Hangzhou, China
| | | | - Catherine A. Taylor
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Dongliang Zhang
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| | - Donald M. McDonald
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, UCSF, San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| |
Collapse
|
5
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025; 9:618-637. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
6
|
Åkesson P, Mellhammar L, Rasmussen M, Inghammar M, Jesperson S, Månsson F, Economou Lundeberg E, Walles J, Wallberg M, Frigyesi A, Linder A. Aerosolized Dornase Alfa (DNase I) for the Treatment of Severe Respiratory Failure in COVID-19: A Randomized Controlled Trial. Open Forum Infect Dis 2025; 12:ofaf246. [PMID: 40365079 PMCID: PMC12069806 DOI: 10.1093/ofid/ofaf246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background Lung injury in COVID-19 is characterized by neutrophil invasion and the release of neutrophil extracellular traps (NETs). An aberrant NET formation may induce local inflammation and increase sputum viscosity. Inhalation of DNase I (dornase alfa) is a treatment option that degrades NETs in the airways. Previous case series have indicated positive clinical effects of inhaled dornase alfa. Methods Patients admitted to the hospital with acute COVID-19 and hypoxia (oxygen saturation <90%) were randomly assigned to receive aerosolized dornase alfa twice daily for 5 days or a placebo in addition to standard of care. The primary outcome was discharge from the hospital or an oxygen saturation >93% without respiratory support. Results In total, 76 patients were randomized. The study was stopped when the Omicron virus variant appeared. The clinical response rate did not differ between patients receiving the active substance and placebo. Secondary outcomes were similar across groups, such as mortality, a new episode of hypoxia, length of stay in the hospital, and adverse events. A subanalysis of patients older or younger than 65 years showed no differences in primary or secondary outcomes. Conclusions Aerosolized dornase alfa failed to improve hypoxia in hospitalized patients with acute COVID-19. The study was conducted during a time of heterogeneity in viral variants and vaccination status of participants. Whether dornase alfa affects the outcomes in other respiratory infections requires further study.
Collapse
Affiliation(s)
- Per Åkesson
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Lisa Mellhammar
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Magnus Rasmussen
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Malin Inghammar
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Infectious Diseases, University Hospital, Lund, Sweden
| | - Sara Jesperson
- Clinical Studies Sweden–Forum South, Skåne University Hospital, Lund, Sweden
| | - Fredrik Månsson
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | | | - John Walles
- Department of Infectious Diseases, Central Hospital of Kristianstad, Kristianstad, Sweden
- Clinical Infection Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Martin Wallberg
- Department of Pulmonary Diseases, Skåne University Hospital, Lund, Sweden
| | - Attila Frigyesi
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Wei S, Dai Z, Wu L, Xiang Z, Yang X, Jiang L, Du Z. Lactate-induced macrophage HMGB1 lactylation promotes neutrophil extracellular trap formation in sepsis-associated acute kidney injury. Cell Biol Toxicol 2025; 41:78. [PMID: 40304798 PMCID: PMC12043764 DOI: 10.1007/s10565-025-10026-6] [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: 10/30/2024] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Neutrophils play a key role in sepsis-associated acute kidney injury (SAKI), a common and life-threatening complication of organ failure. High mobility group box 1 (HMGB1) modulates inflammatory responses and the formation of neutrophil extracellular traps (NETs). The present work aimed to explore whether HMGB1 lactylation promotes NET formation and exacerbates SAKI. METHODS Venous blood samples were collected from healthy volunteers and SAKI patients. A SAKI mouse model was established using the cecal ligation and puncture method. A coculture system of macrophage-derived exosomes and neutrophils was established. Macrophage-derived exosomes were isolated and identified. ELISAs, immunofluorescence staining, coimmunoprecipitation, and Western blotting were utilized to determine protein levels. RESULTS Elevated blood lactate levels were associated with increased HMGB1 levels in patients with SAKI. In mouse models, lactate increased HMGB1 expression, promoted NET formation, and exacerbated SAKI. Lactate stimulated M1 macrophages to secrete exosomes, leading to the accumulation and release of HMGB1 in the cytoplasm. Additionally, lactate promoted HMGB1 lactylation in macrophages, triggering the release of mitochondrial DNA from neutrophils and activating the cyclic GMP‒AMP synthase/stimulator of interferon genes pathway. CONCLUSION This study revealed that lactate-induced HMGB1 lactylation in macrophages plays a role in promoting NET formation in SAKI through the cGAS/STING pathway. These findings suggest that HMGB1 could be a potential target for therapeutic intervention in SAKI.
Collapse
Affiliation(s)
- Siwei Wei
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zijuan Dai
- Department of Anesthesiology, The Fourth Hospital of Changsha (Affiliated Changsha Hospital of Hunan Normal University), Changsha City, Hunan Province, China
| | - Lei Wu
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zhen Xiang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Xiaoxiao Yang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Liubing Jiang
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China
| | - Zhen Du
- Department of Anesthesiology, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), No. 86, Ziyuan Road, Yuhua District, Changsha City, 410007, Hunan Province, China.
| |
Collapse
|
8
|
Traber KE, Mizgerd JP. The Integrated Pulmonary Immune Response to Pneumonia. Annu Rev Immunol 2025; 43:545-569. [PMID: 40036700 DOI: 10.1146/annurev-immunol-082323-031642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Pneumonia is an acute respiratory infection of the lower respiratory tract. The effectiveness of the host immune response determines the severity of infection, or whether pneumonia occurs at all. The lungs house both innate and adaptive immune systems, which integrate their activities to provide host defense that eliminates microbes and prevents lower respiratory infection from becoming severe. Professional immune cells in the lung, like macrophages and lymphocytes, work with lung constituents, like epithelial cells and fibroblasts, to optimize antimicrobial defense. The dynamics of the immune response during infection and the immune components contributing to defense are influenced by prior experiences with respiratory pathogens, remodeling lung immunity in ways that improve responses against subsequent infections. This review covers how innate and adaptive immune activities coordinate inside the lung to provide integrated and effective immune resistance against respiratory pathogens.
Collapse
Affiliation(s)
- Katrina E Traber
- Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA;
| | - Joseph P Mizgerd
- Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA;
- Department of Virology, Immunology, and Microbiology and Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Mohammadi A, De Luca D, Gauda EB. Characteristics, triggers, treatments, and experimental models of neonatal acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2025; 328:L512-L525. [PMID: 39924963 DOI: 10.1152/ajplung.00312.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/20/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
Neonatal acute respiratory distress syndrome (NARDS) is a severe and potentially life-threatening form of lung injury recently defined by the International Neonatal ARDS Consensus. It is marked by extensive lung inflammation and damage to the alveolar epithelium and vascular endothelium. NARDS can be triggered by direct inflammatory exposures, such as pneumonia and aspiration, and indirect exposures, including sepsis, necrotizing enterocolitis, and chorioamnionitis. This review provides clinicians and researchers with the latest insights on NARDS. We adopt a cross-disciplinary approach to discuss the diagnostic criteria, pathobiology, triggers, epidemiology, and treatments of NARDS. In addition, we summarize existing clinical studies and advanced preclinical models that help address current knowledge gaps. Future research should focus on standardizing the Montreux consensus definition of NARDS in preclinical and clinical studies, identifying biomarkers, developing prediction models, and exploring novel therapies for affected infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology and Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, "A. Béclère" Medical Center, Paris - Saclay University Hospitals, APHP, Paris, France
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France
| | - Estelle B Gauda
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology and Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Talwar S, Harker JA, Openshaw PJM, Thwaites RS. Autoimmunity in long COVID. J Allergy Clin Immunol 2025; 155:1082-1094. [PMID: 39956285 DOI: 10.1016/j.jaci.2025.02.005] [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: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Long COVID (also termed postacute sequelae of SARS-CoV-2, or PASC) affects up to 10% of people recovering from infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnosis is hampered by diffuse symptomatology, lack of biomarkers, incomplete understanding of pathogenesis, and lack of validated treatments. In terms of pathogenesis, hypothesized causes include virus persistence, the legacy of endotheliitis and thrombosis, low-grade tissue-based inflammation and/or scarring, perturbation of the host virome/microbiome, or triggering of autoimmunity. Several studies show preexisting and/or de novo production of autoantibodies after infection with SARS-CoV-2, but the persistence of these antibodies and their role in causing long COVID is debated. Here, we review the mechanisms through which autoimmune responses can arise during and after viral infection, focusing on the evidence for B-cell dysregulation and autoantibody production in acute and long COVID.
Collapse
Affiliation(s)
- Shubha Talwar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - James A Harker
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J M Openshaw
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
11
|
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
|
12
|
Deniau B, Ludes PO, Khalifeh-Ballan P, Fenninger L, Kindo M, Collange O, Geny B, Noll E, Azibani F, Mebazaa A, Pottecher J. Transpulmonary LOX-1 Levels Are Predictive of Acute Respiratory Distress Syndrome After Cardiac Surgery: A Proof-of-Concept Study. Biomedicines 2025; 13:800. [PMID: 40299349 PMCID: PMC12024757 DOI: 10.3390/biomedicines13040800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Acute respiratory distress syndrome (ARDS) is a life-threatening condition that frequently complicates high-risk cardiac surgery. We evaluated the circulating levels and transpulmonary gradient of intracellular proteins in patients at risk of developing ARDS after cardiac surgery using large scale-proteomics. Methods: We enrolled sixteen patients undergoing high-risk cardiac surgery, followed by planned ICU admission. Circulating levels of intracellular proteins were measured at the onset of the surgical procedure, at ICU admission (H0), and 24 h (H24) after surgery in blood samples simultaneously drawn from both the pulmonary artery and the left atrium. The primary endpoint was the occurrence of ARDS between ICU admission and the subsequent 48 h. Results: Among the studied proteins, the levels of intracellular lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) were higher at H24 in the pulmonary artery in patients who developed ARDS (6.96; 95% CI [6.83-7.23]) compared to patients who did not (6.48; 95% CI [6.27-6.66]), p-value = 0.016. The transpulmonary gradient of intracellular LOX-1 levels was not significantly different between ARDS and non-ARDS patients at H0 but it was more negative at H24 in ARDS (-0.23; 95% CI [-0.27, -0.14]) than in non-ARDS patients (0.03; 95% CI [-0.14, 0.32]; p-value= 0.031), with a hazard ratio HR = 0.39 (95% CI [0.18-0.86]); p-value= 0.035. The area under the ROC curve of H24 LOX-1 transpulmonary gradient to predict ARDS occurrence was 0.83 (95% CI [0.62-1.00]). Conclusions: The transpulmonary gradient of intracellular LOX-1 levels was negatively associated with the occurrence of ARDS within the first 48 h after high-risk cardiac surgery, suggesting that lung trapping of LOX-1 may be linked to postoperative ARDS.
Collapse
Affiliation(s)
- Benjamin Deniau
- Department of Anaesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France; (B.D.); (A.M.)
- UMR-S 942, INSERM, MASCOT, Paris University, 75018 Paris, France;
- Department of Medicine, Paris Cité University, 75006 Paris, France
- FHU PROMICE, Hôpital Lariboisière 2, rue Ambroise Paré, 75475 Paris, CEDEX 10, France
- INI CRCT, CHRU Brabois, 54500 Vandoeuvre les Nancy, France
| | - Pierre-Olivier Ludes
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Hautepierre Hospital, Strasbourg University Hospital, 67000 Strasbourg, France (P.K.-B.); (L.F.); (E.N.)
| | - Pamela Khalifeh-Ballan
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Hautepierre Hospital, Strasbourg University Hospital, 67000 Strasbourg, France (P.K.-B.); (L.F.); (E.N.)
| | - Luc Fenninger
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Hautepierre Hospital, Strasbourg University Hospital, 67000 Strasbourg, France (P.K.-B.); (L.F.); (E.N.)
| | - Michel Kindo
- Department of CardioVascular Surgery, Nouvel Hôpital Civil, Strasbourg University Hospital, 67000 Strasbourg, France;
- UR 3072, « Mitochondrie, Stress Oxydant et Protection Musculaire », FMTS, FHU Omicare, Faculty of Medicine, Midwifery and Health Sciences, Strasbourg University, 67081 Strasbourg, France; (O.C.); (B.G.)
| | - Olivier Collange
- UR 3072, « Mitochondrie, Stress Oxydant et Protection Musculaire », FMTS, FHU Omicare, Faculty of Medicine, Midwifery and Health Sciences, Strasbourg University, 67081 Strasbourg, France; (O.C.); (B.G.)
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Bernard Geny
- UR 3072, « Mitochondrie, Stress Oxydant et Protection Musculaire », FMTS, FHU Omicare, Faculty of Medicine, Midwifery and Health Sciences, Strasbourg University, 67081 Strasbourg, France; (O.C.); (B.G.)
- Service de Physiologie et d’Explorations Fonctionnelles, Nouvel Hôpital Civil, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Eric Noll
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Hautepierre Hospital, Strasbourg University Hospital, 67000 Strasbourg, France (P.K.-B.); (L.F.); (E.N.)
- UR 3072, « Mitochondrie, Stress Oxydant et Protection Musculaire », FMTS, FHU Omicare, Faculty of Medicine, Midwifery and Health Sciences, Strasbourg University, 67081 Strasbourg, France; (O.C.); (B.G.)
| | - Fériel Azibani
- UMR-S 942, INSERM, MASCOT, Paris University, 75018 Paris, France;
| | - Alexandre Mebazaa
- Department of Anaesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France; (B.D.); (A.M.)
- UMR-S 942, INSERM, MASCOT, Paris University, 75018 Paris, France;
- Department of Medicine, Paris Cité University, 75006 Paris, France
- FHU PROMICE, Hôpital Lariboisière 2, rue Ambroise Paré, 75475 Paris, CEDEX 10, France
- INI CRCT, CHRU Brabois, 54500 Vandoeuvre les Nancy, France
| | - Julien Pottecher
- Department of Anaesthesiology Critical Care and Perioperative Medicine, Hautepierre Hospital, Strasbourg University Hospital, 67000 Strasbourg, France (P.K.-B.); (L.F.); (E.N.)
- UR 3072, « Mitochondrie, Stress Oxydant et Protection Musculaire », FMTS, FHU Omicare, Faculty of Medicine, Midwifery and Health Sciences, Strasbourg University, 67081 Strasbourg, France; (O.C.); (B.G.)
| |
Collapse
|
13
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [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: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
14
|
Sun J, Liu D, Jin S, Li X, Liu G, Li S, Chen F, Qin X, Zhang Y, Jiang F, Chen D, Pang Q, Hu C, Wu Y, Wang Z. Deletion of BTB and CNC Homology 1 Protects Against Staphylococcus aureus-Induced Acute Lung Injury. J Infect Dis 2025; 231:734-745. [PMID: 39171916 DOI: 10.1093/infdis/jiae392] [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: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
BTB and CNC homology 1 (BACH1) plays a crucial role in the pathogenesis of acute lung injury (ALI) caused by gram-negative bacteria. However, its exact mechanisms in Staphylococcus aureus (SA)-induced ALI, a gram-positive bacterial infection, remain incompletely understood. In this study, we generated a BACH1-knockout mouse model (BACH1-/-) to investigate the role of BACH1 and its underlying mechanisms in regulating the development of sepsis-induced acute lung injury (ALI). Elevated levels of BACH1 were observed in both serum samples from septic patients and mouse models. Deletion of BACH1 alleviated ALI symptoms induced by sepsis. In bone marrow-derived macrophages, BACH1 deletion or knockdown suppressed NF-κB p65 phosphorylation and the induction of pro-inflammatory cytokines. Mechanistic studies demonstrated that BACH1 downregulated tumor necrosis factor-alpha-induced protein 3 (TNFAIP3) mRNA expression by binding to its promoter region. These findings uncover inhibiting BACH1 may be a promising therapeutic strategy for treating gram-positive bacteria-induced ALI.
Collapse
Affiliation(s)
- Jiaojiao Sun
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dapeng Liu
- Department of Anesthesia and Perioperative Medicine, Zao Zhuang Municipal Hospital, Zao Zhuang, China
| | - Sihao Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaolin Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Gang Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shengpeng Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaoyun Qin
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yanli Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fengjuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qingfeng Pang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunxiao Hu
- Department of Transplant Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Yaxian Wu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiqiang Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
15
|
Huang X, Qin H, Zhang R, Jia X, Zhao D, Liu F. Neutrophils are involved in the development and outcomes of plastic bronchitis associated with Mycoplasma pneumoniae pneumonia. Respir Res 2025; 26:92. [PMID: 40057734 PMCID: PMC11890523 DOI: 10.1186/s12931-025-03167-z] [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: 06/25/2024] [Accepted: 02/22/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Previous research has demonstrated a notable increase in neutrophil counts among pediatric patients with plastic bronchitis (PB) associated with Mycoplasma pneumoniae pneumonia (MPP). However, the role of neutrophils in MPP-associated PB remains largely elusive. METHODS This is a nested case-control study that enrolled patients diagnosed with MPP who underwent bronchoscopy in our department during the MPP pandemic from September 2023 to January 2024. We conducted an analysis of clinical characteristics, blood samples, bronchoalveolar lavage fluid (BALF), and cast specimens, correlating these factors with the development and outcomes of PB. RESULTS Among the 557 patients with MPP included in the study, 21 (3.8%) developed PB. The peripheral neutrophil count was identified as an independent risk factor for PB (OR = 3.113 [95%CI 1.050-9.224], P = 0.04) and exhibited strong predictive value for the condition (AUC = 0.885 [95%CI 0.796-0.975], P < 0.001). Notably, there was a marked presence of neutrophil infiltration and neutrophil extracellular traps (NETs) formation in the blood, BALF, and cast samples from patients with PB. Furthermore, the levels of neutrophils and NETs correlated significantly with clinical outcomes. CONCLUSION A high level of neutrophils poses a risk for PB and demonstrates strong predictive value for its diagnosis. Neutrophils and NETs are closely linked to the clinical outcomes of PB in patients with MPP.
Collapse
Affiliation(s)
- Xia Huang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Houbing Qin
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Rui Zhang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xinyi Jia
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Feng Liu
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| |
Collapse
|
16
|
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
|
17
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
18
|
Retter A, Singer M, Annane D. "The NET effect": Neutrophil extracellular traps-a potential key component of the dysregulated host immune response in sepsis. Crit Care 2025; 29:59. [PMID: 39905519 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
|
19
|
Zhang G, Zhang K. Screening and Identification of Neutrophil Extracellular Trap-related Diagnostic Biomarkers for Pediatric Sepsis by Machine Learning. Inflammation 2025; 48:212-222. [PMID: 38795170 DOI: 10.1007/s10753-024-02059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/27/2024]
Abstract
Neutrophil extracellular trap (NET) is released by neutrophils to trap invading pathogens and can lead to dysregulation of immune responses and disease pathogenesis. However, systematic evaluation of NET-related genes (NETRGs) for the diagnosis of pediatric sepsis is still lacking. Three datasets were taken from the Gene Expression Omnibus (GEO) database: GSE13904, GSE26378, and GSE26440. After NETRGs and differentially expressed genes (DEGs) were identified in the GSE26378 dataset, crucial genes were identified by using LASSO regression analysis and random forest analysis on the genes that overlapped in both DEGs and NETRGs. These crucial genes were then employed to build a diagnostic model. The diagnostic model's effectiveness in identifying pediatric sepsis across the three datasets was confirmed through receiver operating characteristic curve (ROC) analysis. In addition, clinical pediatric sepsis samples were collected to measure the expression levels of important genes and evaluate the diagnostic model's performance using qRT-PCR in identifying pediatric sepsis in actual clinical samples. Next, using the CIBERSORT database, the relationship between invading immune cells and diagnostic markers was investigated in more detail. Lastly, to evaluate NET formation, we measured myeloperoxidase (MPO)-DNA complex levels using ELISA. A group of five important genes (MME, BST1, S100A12, FCAR, and ALPL) were found among the 13 DEGs associated with NET formation and used to create a diagnostic model for pediatric sepsis. Across all three cohorts, the sepsis group had consistently elevated expression levels of these five critical genes as compared to the normal group. Area under the curve (AUC) values of 1, 0.932, and 0.966 indicate that the diagnostic model performed exceptionally well in terms of diagnosis. Notably, when applied to the clinical samples, the diagnostic model also showed good diagnostic capacity with an AUC of 0.898, outperforming the effectiveness of traditional inflammatory markers such as PCT, CRP, WBC, and NEU%. Lastly, we discovered that children with high ratings for sepsis also had higher MPO-DNA complex levels. In conclusion, the creation and verification of a five-NETRGs diagnostic model for pediatric sepsis performs better than established markers of inflammation.
Collapse
Affiliation(s)
- Genhao Zhang
- Department of Blood Transfusion, Zhengzhou University First Affiliated Hospital, Zhengzhou, China.
| | - Kai Zhang
- Department of Medical Laboratory, Zhengzhou University Third Affiliated Hospital, Zhengzhou, China
| |
Collapse
|
20
|
He W, Yan L, Hu D, Hao J, Liou Y, Luo G. Neutrophil heterogeneity and plasticity: unveiling the multifaceted roles in health and disease. MedComm (Beijing) 2025; 6:e70063. [PMID: 39845896 PMCID: PMC11751288 DOI: 10.1002/mco2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Neutrophils, the most abundant circulating leukocytes, have long been recognized as key players in innate immunity and inflammation. However, recent discoveries unveil their remarkable heterogeneity and plasticity, challenging the traditional view of neutrophils as a homogeneous population with a limited functional repertoire. Advances in single-cell technologies and functional assays have revealed distinct neutrophil subsets with diverse phenotypes and functions and their ability to adapt to microenvironmental cues. This review provides a comprehensive overview of the multidimensional landscape of neutrophil heterogeneity, discussing the various axes along which diversity manifests, including maturation state, density, surface marker expression, and functional polarization. We highlight the molecular mechanisms underpinning neutrophil plasticity, focusing on the complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications that shape neutrophil responses. Furthermore, we explore the implications of neutrophil heterogeneity and plasticity in physiological processes and pathological conditions, including host defense, inflammation, tissue repair, and cancer. By integrating insights from cutting-edge research, this review aims to provide a framework for understanding the multifaceted roles of neutrophils and their potential as therapeutic targets in a wide range of diseases.
Collapse
Affiliation(s)
- Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Dongxue Hu
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| |
Collapse
|
21
|
Grudzinska F, Faniyi AA, Belchamber KBR, Chen C, Stockley R, Jasper A, Parekh D, Sapey E, Scott A, Thickett DR. Hospitalised older adults with community-acquired pneumonia and sepsis have dysregulated neutrophil function but preserved glycolysis. Thorax 2025; 80:97-104. [PMID: 39689942 PMCID: PMC11877105 DOI: 10.1136/thorax-2024-222215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE Community-acquired pneumonia (CAP) is a leading cause of hospitalisation in older adults and is associated with a high likelihood of adverse outcomes. Given the ageing population and lack of therapeutic advances in CAP, new strategies to manage the burden of this disease are needed. Neutrophil dysfunction has been widely demonstrated in CAP and is associated with poor outcomes. We hypothesised that impaired glycolytic metabolism was driving neutrophil dysfunction in older adults with CAP. METHODS To investigate the mechanism underlying neutrophil dysfunction in CAP, we recruited older adults with CAP and sepsis, age-matched controls and healthy young adults to assess neutrophil function and glycolytic metabolism in peripheral blood neutrophils. RESULTS We demonstrate that neutrophils from older donors with CAP display a broad range of functional defects, including inaccurate migration to interleukin 8, impaired respiratory burst in response to phorbol 12-myristate 13-acetate and increased spontaneous degranulation compared with age-matched controls. Glycolysis (assessed by extracellular flux and RNA-sequencing) was not significantly altered between age-matched groups; however, basal rates of neutrophil glycolysis were significantly higher in patients with CAP and older adult controls compared with healthy young adults, and stimulated glycolysis was significantly higher in young adults compared with older adults with and without CAP. CONCLUSIONS Our findings suggest that neutrophil dysfunction in older adults with CAP may be implicated in poor outcomes, irrespective of glycolytic metabolism.
Collapse
Affiliation(s)
- Frances Grudzinska
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- School of Translational Medicine, University of Nottingham, Nottingham, UK
| | - Aduragbemi A Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Kylie B R Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- The University of Manchester Maternal and Fetal Health Research Centre, Manchester, UK
| | - Celine Chen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Robert Stockley
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Alice Jasper
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- NIHR Birmingham Clinical Research Facility, Birmingham, UK
| | - Elizabeth Sapey
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- PIONEER HDR-UK Hub in Acute Care, Birmingham, UK
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
| | - David R Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, Birmingham, UK
| |
Collapse
|
22
|
Englert H, Rangaswamy C, Kullik GA, Divivier M, Göbel J, Hermans‐Borgmeyer I, Borgmeyer U, Mowen KA, Beerens M, Frye M, Mailer RK, Gelderblom M, Stavrou EX, Preston RJS, Schneider SW, Fuchs TA, Renné T. Sepsis-induced NET formation requires MYD88 but is independent of GSDMD and PAD4. FASEB J 2025; 39:e70301. [PMID: 39777764 PMCID: PMC11707982 DOI: 10.1096/fj.202402514r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025]
Abstract
Neutrophils are peripheral blood-circulating leukocytes that play a pivotal role in host defense against bacterial pathogens which upon activation, they release web-like chromatin structures called neutrophil extracellular traps (NETs). Here, we analyzed and compared the importance of myeloid differentiation factor 88 (MYD88), peptidyl arginine deiminase 4 (PAD4), and gasdermin D (GSDMD) for NET formation in vivo following sepsis and neutrophilia challenge. Injection of lipopolysaccharide (LPS)/E. coli or the transgenic expression of granulocyte colony-stimulating factor (G-CSF), each induced NET-mediated lethal vascular occlusions in mice with combined genetic deficiency in Dnase1 and Dnase1l3 (D1/D1l3-/-). In accordance with the signaling of toll-like receptors, Myd88/D1/D1l3-/- animals were protected from the formation of lethal intravascular NETs during septic conditions. However, this protection was not observed during neutrophilia. It was unexpected to find that both Gsdmd/D1/D1l3-/- and Pad4/D1/D1l3-/- mice were fully capable of forming NETs upon LPS/E.coli challenge. Sepsis equally triggered a similar inflammatory response in these mice characterized by formation of DNA-rich thrombi, vessel occlusions, and mortality from pulmonary embolism, compared to D1/D1l3-/- mice. Pharmacologic GSDMD inhibitors did not reduce PMA-stimulated NET formation in ex vivo models either. Similarly, neither Pad4 nor GSDMD deficiency affected intravascular occlusive NET formation upon neutrophilia challenge. The magnitude of NET production, multi-organ damage, and lethality were comparable to those observed in challenged control mice. In conclusion, our data indicate that NET formation during experimental sepsis and neutrophilia is regulated by distinct stimulus-dependent pathways that may be independent of canonical PAD4 and GSDMD.
Collapse
Affiliation(s)
- Hanna Englert
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Giuliano A. Kullik
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mylène Divivier
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Josephine Göbel
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Irm Hermans‐Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Uwe Borgmeyer
- Transgenic Mouse Unit, Center for Molecular Neurobiology HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Kerri A. Mowen
- Chemical PhysiologyThe Scripps InstituteLa JollaCaliforniaUSA
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, KielHamburgGermany
| | - Reiner K. Mailer
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Mathias Gelderblom
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Evi X. Stavrou
- Medicine Service, Section of Hematology‐OncologyLouis Stokes Veterans Administration Medical CenterClevelandOhioUSA
- Department of Medicine, Hematology and Oncology DivisionCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Roger J. S. Preston
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Stefan W. Schneider
- Department of Dermatology and VenereologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tobias A. Fuchs
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- NeutrolisCambridgeMassachusettsUSA
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
- Center for Thrombosis and Hemostasis (CTH)Johannes Gutenberg University Medical CenterMainzGermany
| |
Collapse
|
23
|
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
|
24
|
Karasu E, Halbgebauer R, Schütte L, Greven J, Bläsius FM, Zeller J, Winninger O, Braig D, Messerer DAC, Berger B, Feuerstein H, Schultze A, Peter K, Knippschild U, Horst K, Hildebrand F, Eisenhardt SU, Huber-Lang M. A conformational change of C-reactive protein drives neutrophil extracellular trap formation in inflammation. BMC Biol 2025; 23:4. [PMID: 39773175 PMCID: PMC11708171 DOI: 10.1186/s12915-024-02093-8] [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: 10/25/2023] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND C-reactive protein (CRP) represents a routine diagnostic marker of inflammation. Dissociation of native pentameric CRP (pCRP) into the monomeric structure (mCRP) liberates proinflammatory features, presumably contributing to excessive immune cell activation via unknown molecular mechanisms. RESULTS In a multi-translational study of systemic inflammation, we found a time- and inflammation-dependent pCRP dissociation into mCRP. We were able to confirm that mCRP co-localizes with leukocytes at the site of injury after polytrauma and therefore assessed whether the CRP conformation potentiates neutrophil activation. We found mCRP-induced neutrophil-extracellular trap formation in vitro and ex vivo involving nicotinamide adenine dinucleotide phosphate oxidase activation, p38/mitogen-activated protein kinase signaling, and histone H3 citrullination. Mimicking the trauma milieu in a human ex vivo whole blood model, we found significant mCRP generation as well as NET formation, prevented by blocking pCRP conformational changes. CONCLUSIONS Our data provide novel molecular insights how CRP dissociation contributes to neutrophil activation as driver of various inflammatory disorders.
Collapse
Affiliation(s)
- Ebru Karasu
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Lena Schütte
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Johannes Greven
- Department of Orthopedic Trauma Surgery, RWTH Aachen University, Aachen, Germany
| | - Felix M Bläsius
- Department of Orthopedic Trauma Surgery, RWTH Aachen University, Aachen, Germany
| | - Johannes Zeller
- Department of Plastic and Hand Surgery, Medical Faculty of the University of Freiburg, University of Freiburg Medical Centre Freiburg, Freiburg, Germany
| | - Oscar Winninger
- Department of Plastic and Hand Surgery, Medical Faculty of the University of Freiburg, University of Freiburg Medical Centre Freiburg, Freiburg, Germany
| | - David Braig
- Department of Plastic and Hand Surgery, Medical Faculty of the University of Freiburg, University of Freiburg Medical Centre Freiburg, Freiburg, Germany
| | | | - Bettina Berger
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Hendrik Feuerstein
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Anke Schultze
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Karlheinz Peter
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Medical Center, 89081, Ulm, Germany
| | - Klemens Horst
- Department of Orthopedic Trauma Surgery, RWTH Aachen University, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopedic Trauma Surgery, RWTH Aachen University, Aachen, Germany
| | - Steffen U Eisenhardt
- Department of Plastic and Hand Surgery, Medical Faculty of the University of Freiburg, University of Freiburg Medical Centre Freiburg, Freiburg, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany.
| |
Collapse
|
25
|
Tan L, Zhang C, Kou X, Zhao L, Wu D, Li J, Yu C, Xu T, Gao L, Mao X, Zhao C. Apoptotic Vesicles Attenuate Acute Lung Injury via CD73-Mediated Inhibition of Platelet Activation and NETosis. Int J Nanomedicine 2025; 20:91-107. [PMID: 39802376 PMCID: PMC11717653 DOI: 10.2147/ijn.s485012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is a life-threatening type of acute lung injury (ALI) characterized by elevated mortality rates and long-term effects. To date, no pharmacological treatment has proven effective for ARDS. Mesenchymal stem cell-derived apoptotic vesicles (apoVs) were recently found to have excellent therapeutic potential for inflammatory diseases. In this study, our aim was to investigate the therapeutic effects and underlying mechanisms of apoVs in ALI. Methods ALI was induced in mice through intratracheal instillation of lipopolysaccharide (LPS). ApoVs were then administered two hours post-induction, and their impacts on platelet activation, neutrophil infiltration, and NETosis were assessed. Additionally, the role of CD73 in mediating these effects was thoroughly investigated. Results ApoVs inhibit platelet activation, thereby impeding the infiltration of neutrophils into the lung and the initiation of NETosis, ultimately alleviating ALI. Remarkably, apoVs were enriched with CD73, which was critical for apoV-mediated repression of platelet activation and neutrophil NETosis, as well as the therapeutic effects observed in lung injury. Conclusion This study reveals that apoVs inhibit platelet activity and neutrophil NETosis via CD73, offering an innovative and effective cell-free therapeutic strategy for ALI/ARDS.
Collapse
Affiliation(s)
- Lingping Tan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Chi Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, People’s Republic of China
| | - Lu Zhao
- Department of Orthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, People’s Republic of China
| | - Di Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, People’s Republic of China
| | - Jinyu Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Chuanying Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Tansi Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Li Gao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, People’s Republic of China
| | - Chuanjiang Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| |
Collapse
|
26
|
Wang Y, Guo J, Zhang D, Shi C, Zhang X, Gong Z. IDH1/MDH1 deacetylation promotes NETosis by regulating OPA1 and autophagy. Int Immunopharmacol 2024; 143:113270. [PMID: 39353390 DOI: 10.1016/j.intimp.2024.113270] [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: 08/15/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND As a heterogeneous and life-threatening disease, the pathogenesis of acute liver failure (ALF) is complex. Our previous study has shown that IDH1/MDH1 deacetylation promotes ALF by regulating NETosis (a novel mode of cell death). In this article, we explore the manners of IDH1/MDH1 deacetylation regulates NETosis. METHODS In vitro experiments, the formation of NETs was detected by immunofluorescence staining and Western blotting. LC3 fluorescence staining was used to detect autophagosome formation. To observe mitochondrial morphology, cells were stained by Mito-Tracker Red. Western blotting was used to detect the levels of autophagy protein and mitochondrial dynamin. In vivo experiments, the ALF model in mouse was established with LPS/D-gal, and the formation of NETs was detected by immunofluorescence staining and Western blotting. The autophagy levels were detected by Western blotting in liver samples. RESULTS In dHL-60 cells, Western blotting results showed that the expression of OPA1 was higher in the IDH1/MDH1 deacetylated group compared with the IDH1/MDH1 WT group. And histone deacetylase inhibitor 6 (HDAC6i, ACY1215) decreased the expression level of OPA1 in IDH1/MDH1 deacetylated group. IDH1/MDH1 deacetylation increased the expression levels of both LC3B-II and Beclin 1, while decreasing the expression level of P62. It was reversed by ACY1215. Combined with our previous experiments, IDH1/MDH1 deacetylation upregulated autophagy concomitant with the increased expression of the markers of NETs formation. In a mouse model of ALF, ACY1215 further decreased the expression levels of LC3B-II and Beclin 1, while increasing the expression level of P62 in IDH1/MDH1 deacetylated mice. CONCLUSIONS IDH1/MDH1 deacetylation promoted NETosis by regulating autophagy and OPA1 in vitro. The regulation of neutrophil autophagy on NETosis during IDH1/MDH1 deacetylation might be masked in mice. ACY1215 might attenuate NETosis by regulating neutrophil autophagy, which alleviated ALF aggravated by IDH1/MDH1 deacetylation.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xiaoya Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
27
|
Cheng S, Li Y, Sun X, Liu Z, Guo L, Wu J, Yang X, Wei S, Wu G, Xu S, Yang F, Wu J. The impact of glucose metabolism on inflammatory processes in sepsis-induced acute lung injury. Front Immunol 2024; 15:1508985. [PMID: 39712019 PMCID: PMC11659153 DOI: 10.3389/fimmu.2024.1508985] [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: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Acute lung injury (ALI) is a prevalent and critical complication of sepsis, marked by high incidence and mortality rates, with its pathogenesis still not being fully elucidated. Recent research has revealed a significant correlation between the metabolic reprogramming of glucose and sepsis-associated ALI (S-ALI). Throughout the course of S-ALI, immune cells, including macrophages and dendritic cells, undergo metabolic shifts to accommodate the intricate demands of immune function that emerge as sepsis advances. Indeed, glucose metabolic reprogramming in S-ALI serves as a double-edged sword, fueling inflammatory immune responses in the initial stages and subsequently initiating anti-inflammatory responses as the disease evolves. In this review, we delineate the current research progress concerning the pathogenic mechanisms linked to glucose metabolic reprogramming in S-ALI, with a focus on the pertinent immune cells implicated. We encapsulate the impact of glucose metabolic reprogramming on the onset, progression, and prognosis of S-ALI. Ultimately, by examining key regulatory factors within metabolic intermediates and enzymes, We have identified potential therapeutic targets linked to metabolic reprogramming, striving to tackle the inherent challenges in diagnosing and treating Severe Acute Lung Injury (S-ALI) with greater efficacy.
Collapse
Affiliation(s)
- Shilei Cheng
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Yufei Li
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine (TCM), Jinan, China
| | - Xiaoliang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhirui Liu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Liang Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jueheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaohan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Sisi Wei
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Guanghan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Shilong Xu
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Fan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| |
Collapse
|
28
|
Mittendorfer M, Pierre L, Huzevka T, Schofield J, Abrams ST, Wang G, Toh CH, Bèchet NB, Caprnja I, Kjellberg G, Aswani A, Olm F, Lindstedt S. Restoring discarded porcine lungs by ex vivo removal of neutrophil extracellular traps. J Heart Lung Transplant 2024; 43:1919-1929. [PMID: 39038563 DOI: 10.1016/j.healun.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND By causing inflammation and tissue damage, neutrophil extracellular traps (NETs) constitute an underlying mechanism of aspiration-induced lung injury, a major factor of the low utilization of donor lungs in lung transplantation (LTx). METHODS To determine whether NET removal during ex vivo lung perfusion (EVLP) can restore lung function and morphology in aspiration-damaged lungs, gastric aspiration lung injury was induced in 12 pigs. After confirmation of acute respiratory distress syndrome, the lungs were explanted and assigned to NET removal connected to EVLP (treated) (n = 6) or EVLP only (nontreated) (n = 6). Hemodynamic measurements were taken, and blood and tissue samples were collected to assess lung function, morphology, levels of cell-free DNA, extracellular histones, and nucleosomes as markers of NETs, as well as cytokine levels. RESULTS After EVLP and NET removal in porcine lungs, PaO2/FiO2 ratios increased significantly compared to those undergoing EVLP alone (p = 0.0411). Treated lungs had lower cell-free DNA (p = 0.0260) and lower levels of extracellular histones in EVLP perfusate (p= 0.0260) than nontreated lungs. According to histopathology, treated lungs showed less immune cell infiltration and less edema compared with nontreated lungs, which was reflected in decreased levels of proinflammatory cytokines in EVLP perfusate and bronchoalveolar lavage fluid. CONCLUSIONS To conclude, removing NETs during EVLP improved lung function and morphology in aspiration-damaged donor lungs. The ability to remove NETs during EVLP could represent a new therapeutic approach for LTx and potentially expand the donor pool for transplantation.
Collapse
Affiliation(s)
- Margareta Mittendorfer
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Leif Pierre
- Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Tibor Huzevka
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Jeremy Schofield
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom; Roald Dahl Haemostasis & Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Nicholas B Bèchet
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Ilma Caprnja
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Gunilla Kjellberg
- Department of Thoracic Surgery and Anaesthesiology, Uppsala University Hospital, Uppsala, Sweden
| | - Andrew Aswani
- Department of Critical Care, Guy's and St Thomas's NHS Foundation Trust, London, United Kingdom; Santersus AG, Zurich, Switzerland
| | - Franziska Olm
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Cardiothoracic Surgery and Transplantation, Lund University Hospital, Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden; Lund Stem Cell Centre, Lund University, Lund, Sweden.
| |
Collapse
|
29
|
Priya K, Thacker H, Chaubey M, Rai M, Singh S, Rawat S, Giri K, Mohanty S, Rai G. Dexamethasone and IFN-γ primed mesenchymal stem cells conditioned media immunomodulates aberrant NETosis in SLE via PGE2 and IDO. Front Immunol 2024; 15:1461841. [PMID: 39544931 PMCID: PMC11560778 DOI: 10.3389/fimmu.2024.1461841] [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: 07/09/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Background Systemic Lupus Erythematosus (SLE) is characterized by dysregulated immune responses, with neutrophil extracellular traps (NETs) playing a significant role. NETs are recognized by autoantibodies in SLE patients, exacerbating pathology. Both excessive NET formation and impaired degradation contribute to SLE pathophysiology. Objective To investigate the immunomodulatory effects of Dexamethasone-primed Wharton's jelly (WJ) derived MSCs CM (DW) and IFN-γ-primed WJ-MSCs-CM (IW) on NETosis and associated protein markers in SLE patients' LPS or ribonucleoprotein immune complexes (RNP ICs) induced neutrophils and in pristane induced lupus (PIL) model. And to elucidate the mechanism involved therein. Methods We investigated the immunomodulatory effects of DW and IW on NETosis in SLE. Utilizing ex vivo and in vivo models, we assessed the impact of preconditioned media on NET formation and associated protein markers neutrophil elastase (NE), citrullinated histone (citH3), myeloperoxidase (MPO), cytoplasmic and mitochondrial ROS production. We also examined the involvement of key immunomodulatory factors present in DW and IW, including prostaglandin E2 (PGE2), indoleamine 2,3-dioxygenase (IDO), and transforming growth factor-beta (TGF-β). Results Preconditioned media effectively suppressed NETosis and reduced ROS generation in SLE neutrophils, indicating their immunomodulatory potential. Inhibition studies implicated IDO and PGE2 in mediating this effect. Combined treatment with DW or IW together with hydroxychloroquine (HCQ) demonstrated superior efficacy over HCQ alone, a standard SLE medication. In PIL mouse model, DW and IW treatments reduced NETosis, ROS generation, as evidenced by decreased NET-associated protein expression in vital organs. Conclusion Our study highlights the multifaceted impact of IW and DW on NETosis, ROS dynamics, and lupus severity in SLE. These findings underscore the potential of preconditioned media for the development of targeted, personalized approaches for SLE treatment.
Collapse
Affiliation(s)
- Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Hiral Thacker
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Manaswi Chaubey
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Madhukar Rai
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, India
| | - Kiran Giri
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
30
|
Wick KD, Ware LB, Matthay MA. Acute respiratory distress syndrome. BMJ 2024; 387:e076612. [PMID: 39467606 DOI: 10.1136/bmj-2023-076612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The understanding of acute respiratory distress syndrome (ARDS) has evolved greatly since it was first described in a 1967 case series, with several subsequent updates to the definition of the syndrome. Basic science advances and clinical trials have provided insight into the mechanisms of lung injury in ARDS and led to reduced mortality through comprehensive critical care interventions. This review summarizes the current understanding of the epidemiology, pathophysiology, and management of ARDS. Key highlights include a recommended new global definition of ARDS and updated guidelines for managing ARDS on a backbone of established interventions such as low tidal volume ventilation, prone positioning, and a conservative fluid strategy. Future priorities for investigation of ARDS are also highlighted.
Collapse
Affiliation(s)
- Katherine D Wick
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lorraine B Ware
- Departments of Medicine and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
31
|
Liang P, Zhu M, Sun X, Wang L, Li B, Ming S, Younis M, Yang J, Wu Y, Huang X. LncRNA-mRNA co-expression analysis reveals aquaporin-9-promoted neutrophil extracellular trap formation and inflammatory activation in sepsis. Int Immunopharmacol 2024; 140:112916. [PMID: 39133961 DOI: 10.1016/j.intimp.2024.112916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection. However, the precise regulatory mechanism of sepsis remains unclear. Using a strand-specific RNA-sequencing, we identified 115 hub differentially expressed long noncoding RNAs (lncRNAs) and 443 mRNAs in septic patients, primarily participated in crucial pathways including neutrophil extracellular trap (NET) formation and toll-like receptor signaling. Notably, NETs related gene aquaporin-9 (AQP9) and its associated lncRNAs exhibited significant upregulation in septic neutrophils. Functional experiments revealed AQP9 interacts with its lncRNAs to augment the formation of neutrophil NETs. In murine sepsis models, AQP9 inhibition with phloretin reduced proinflammatory cytokine production and lung damage. These findings provide crucial insights into the regulatory role of AQP9 in sepsis, unraveling its interaction with associated lncRNAs in transmitting downstream signals, holding promise in informing the development of novel therapeutic strategies aimed at ameliorating the debilitating effects of sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 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, Guangdong 519000, China
| | - Manman Zhu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Perinatal Medical Center, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Muhammad Younis
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Jianhua Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 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, Guangdong 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 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, Guangdong 519000, China.
| |
Collapse
|
32
|
Li J, Yang D, Lin L, Yu L, Chen L, Lu K, Lan J, Zeng Y, Xu Y. Important functions and molecular mechanisms of aquaporins family on respiratory diseases: potential translational values. J Cancer 2024; 15:6073-6085. [PMID: 39440058 PMCID: PMC11493008 DOI: 10.7150/jca.98829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/25/2024] [Indexed: 10/25/2024] Open
Abstract
Aquaporins (AQPs) are a subgroup of small transmembrane transporters that are distributed in various types of tissues, including the lung, kidney, heart and central nervous system. It is evident that respiratory diseases represent a significant global health concern, with a considerable number of deaths occurring worldwide. Recent researches have demonstrated that AQPs play a pivotal role in respiratory diseases, including chronic obstructive pulmonary disease (COPD), asthma, acute respiratory distress syndrome (ARDS), and particularly non-small cell lung cancer (NSCLC). In the context of NSCLC, the overexpression of AQP1, AQP3, AQP4, and AQP5 has been demonstrated to facilitate tumor angiogenesis, as well as the proliferation, migration, and invasiveness of tumor cells. This review concisely explores the role of AQP family on respiratory diseases, to assess their clinical and translational significance for understanding molecular pathogenesis. However, the potential translation of AQPs biomarkers into clinical applications is promising and the understanding of the precise mechanisms influencing respiratory diseases is still ongoing. Addressing the challenges and outlining the future perspectives in AQPs development is essential for clinical progress in a concise manner.
Collapse
Affiliation(s)
- Jinshan Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Dongyong Yang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Lanlan Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Luyang Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Kaiqiang Lu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Jieli Lan
- Clinical Research Unit, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Yuan Xu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| |
Collapse
|
33
|
Moussavi-Harami SF, Cleary SJ, Magnen M, Seo Y, Conrad C, English BC, Qiu L, Wang KM, Abram CL, Lowell CA, Looney MR. Neutrophil-specific Shp1 loss results in lethal pulmonary hemorrhage in mouse models of acute lung injury. J Clin Invest 2024; 134:e183161. [PMID: 39352872 PMCID: PMC11645157 DOI: 10.1172/jci183161] [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: 05/23/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with significant morbidity and mortality, and neutrophils are critical to its pathogenesis. Neutrophil activation is closely regulated by inhibitory tyrosine phosphatases including Src homology region 2 domain-containing phosphatase-1 (Shp1). Here, we report that loss of neutrophil Shp1 in mice produced hyperinflammation and lethal pulmonary hemorrhage in sterile inflammation and pathogen-induced models of acute lung injury (ALI) through a Syk kinase-dependent mechanism. We observed large intravascular neutrophil clusters, perivascular inflammation, and excessive neutrophil extracellular traps in neutrophil-specific Shp1-KO mice, suggesting an underlying mechanism for the observed pulmonary hemorrhage. Targeted immunomodulation through the administration of a Shp1 activator (SC43) reduced agonist-induced reactive oxygen species in vitro and ameliorated ALI-induced alveolar neutrophilia and NETs in vivo. We propose that the pharmacologic activation of Shp1 has the potential to fine tune neutrophil hyperinflammation that is central to the pathogenesis of ARDS.
Collapse
Affiliation(s)
- S. Farshid Moussavi-Harami
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Division of Pediatric Critical Care Medicine, Department of Pediatrics
| | - Simon J Cleary
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Mélia Magnen
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Yurim Seo
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Catharina Conrad
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | | | - Longhui Qiu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Kristin M. Wang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Clare L. Abram
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | | | - Mark R. Looney
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
34
|
Kim HI, Han Y, Kim MH, Boo M, Cho KJ, Kim HL, Lee IS, Jung JH, Kim W, Um JY, Park J, Ko SG. The multi-herbal decoction SH003 alleviates LPS-induced acute lung injury by targeting inflammasome and extracellular traps in neutrophils. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155926. [PMID: 39128302 DOI: 10.1016/j.phymed.2024.155926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/07/2024] [Accepted: 07/28/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is a devastating condition caused by sepsis, pneumonia, trauma, and more recently, COVID-19. SH003, an herbal formula consisted of Astragalus membranaceus, Angelica gigas and Trichosanthes kirilowii, is known for its effects on cancer and immunoregulation. HYPOTHESIS/PURPOSE Previous studies show SH003 exerts a promising anti-inflammatory effect. This study investigates the effect of modified SH003 on ALI using in silico, in vivo, and in vitro models. STUDY DESIGN AND METHODS We performed in silico-based analysis of SH003 on ALI-related pathways. C57BL/6 mice were intraperitoneally subjected to lipopolysaccharide (LPS) to induce septic ALI, followed by oral administration of SH003 for 2 weeks. Dexamethasone was used as the positive control. Human peripheral blood-derived polymorphonuclear neutrophils (PMN) were used to investigate the effect and mechanisms of SH003 on neutrophil extracellular trap (NET) formation. RESULTS Network pharmacology analysis suggested SH003 regulates lung inflammation by modulating NET formation. SH003 significantly reduced mortality in sepsis in vivo by inhibiting local and systemic inflammation, likely via nuclear factor kappa B and mitogen-activated protein kinase pathways-mediated inflammasome suppression. SH003 also decreased NET-related markers in lung tissues and inhibited LPS- and phorbol myristate acetate-induced NET formation in PMN. Cytometry time-of-flight analysis confirmed regulation of NETosis-related pathways by SH003. CONCLUSION SH003 effectively inhibits excessive immune responses in the lung by suppressing inflammasome activation and NET formation. These findings suggest SH003 as a potential therapeutic agent for septic ALI.
Collapse
Affiliation(s)
- Hyo In Kim
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Yohan Han
- Department of Microbiology and Sarcopenia Total Solution Center, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Mi-Hye Kim
- College of Korean Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Mina Boo
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang-Jin Cho
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hye-Lin Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - In-Seon Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Hoon Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Woojin Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinbong Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, Kyung Hee University, Seoul, Republic of Korea.
| | - Seong-Gyu Ko
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Rodriguez Moore G, Melo-Escobar I, Stegner D, Bracko O. One immune cell to bind them all: platelet contribution to neurodegenerative disease. Mol Neurodegener 2024; 19:65. [PMID: 39334369 PMCID: PMC11438031 DOI: 10.1186/s13024-024-00754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease (AD) and related dementias (ADRD) collectively affect a significant portion of the aging population worldwide. The pathological progression of AD involves not only the classical hallmarks of amyloid beta (Aβ) plaque buildup and neurofibrillary tangle development but also the effects of vasculature and chronic inflammatory processes. Recently, platelets have emerged as central players in systemic and neuroinflammation. Studies have shown that patients with altered platelet receptor expression exhibit accelerated cognitive decline independent of traditional risk factors. Additionally, platelets from AD patients exhibit heightened unstimulated activation compared to control groups. Platelet granules contain crucial AD-related proteins like tau and amyloid precursor protein (APP). Dysregulation of platelet exocytosis contributes to disease phenotypes characterized by increased bleeding, stroke, and cognitive decline risk. Recent studies have indicated that these effects are not associated with the quantity of platelets present in circulation. This underscores the hypothesis that disruptions in platelet-mediated inflammation and healing processes may play a crucial role in the development of ADRD. A thorough look at platelets, encompassing their receptors, secreted molecules, and diverse roles in inflammatory interactions with other cells in the circulatory system in AD and ADRD, holds promising prospects for disease management and intervention. This review discusses the pivotal roles of platelets in ADRD.
Collapse
Affiliation(s)
| | - Isabel Melo-Escobar
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA.
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
36
|
Cleary SJ, Qiu L, Seo Y, Baluk P, Liu D, Serwas NK, Cyster JG, McDonald DM, Krummel MF, Looney MR. Intravital imaging of pulmonary lymphatics in inflammation and metastatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612619. [PMID: 39345499 PMCID: PMC11430110 DOI: 10.1101/2024.09.12.612619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Intravital microscopy has enabled the study of immune dynamics in the pulmonary microvasculature, but many key events remain unseen because they occur in deeper lung regions. We therefore developed a technique for stabilized intravital imaging of bronchovascular cuffs and collecting lymphatics surrounding pulmonary veins in mice. Intravital imaging of pulmonary lymphatics revealed ventilation-dependence of steady-state lung lymph flow and ventilation-independent lymph flow during inflammation. We imaged the rapid exodus of migratory dendritic cells through lung lymphatics following inflammation and measured effects of pharmacologic and genetic interventions targeting chemokine signaling. Intravital imaging also captured lymphatic immune surveillance of lung-metastatic cancers and lymphatic metastasis of cancer cells. To our knowledge, this is the first imaging of lymph flow and leukocyte migration through intact pulmonary lymphatics. This approach will enable studies of protective and maladaptive processes unfolding within the lungs and in other previously inaccessible locations.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, CA, USA
| | - Dan Liu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, UCSF, CA, USA
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, Zhejiang, China
| | | | - Jason G. Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, UCSF, CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| | - Donald M. McDonald
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, UCSF, CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
- Bakar ImmunoX Initiative, UCSF, CA, USA
| |
Collapse
|
37
|
Zhang M, Li S, Ying J, Qu Y. Neutrophils: a key component in ECMO-related acute organ injury. Front Immunol 2024; 15:1432018. [PMID: 39346902 PMCID: PMC11427252 DOI: 10.3389/fimmu.2024.1432018] [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: 05/13/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Extracorporeal membrane oxygenation (ECMO), as an extracorporeal life support technique, can save the lives of reversible critically ill patients when conventional treatments fail. However, ECMO-related acute organ injury is a common complication that increases the risk of death in critically ill patients, including acute kidney injury, acute brain injury, acute lung injury, and so on. In ECMO supported patients, an increasing number of studies have shown that activation of the inflammatory response plays an important role in the development of acute organ injury. Cross-cascade activation of the complement system, the contact system, and the coagulation system, as well as the mechanical forces of the circuitry are very important pathophysiological mechanisms, likely leading to neutrophil activation and the production of neutrophil extracellular traps (NETs). NETs may have the potential to cause organ damage, generating interest in their study as potential therapeutic targets for ECMO-related acute organ injury. Therefore, this article comprehensively summarized the mechanism of neutrophils activation and NETs formation following ECMO treatment and their actions on acute organ injury.
Collapse
Affiliation(s)
- Mingfu Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Xue GZ, Ma HZ, Wuren TN. The role of neutrophils in chronic cough. Hum Cell 2024; 37:1316-1324. [PMID: 38913146 DOI: 10.1007/s13577-024-01089-4] [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: 02/22/2024] [Accepted: 05/18/2024] [Indexed: 06/25/2024]
Abstract
Chronic cough is a common disorder lasting more than 8 weeks and affecting all age groups. The evidence supporting the role of neutrophils in chronic cough pathology is based on many patients with chronic cough developing airway neutrophilia. How neutrophils influence the development of chronic cough is unknown. However, they are likely involved in multiple aspects of cough etiology, including promoting airway inflammation, airway remodeling, hyper-responsiveness, local neurogenic inflammation, and other possible mechanisms. Neutrophilic airway inflammation is also associated with refractory cough, poor control of underlying diseases (e.g., asthma), and insensitivity to cough suppressant therapy. The potential for targeting neutrophils in chronic cough needs exploration, including developing new drugs targeting one or more neutrophil-mediated pathways or altering the neutrophil phenotype to alleviate chronic cough. How the airway microbiome differs, plays a role, and interacts with neutrophils in different cough etiologies is poorly understood. Future studies should focus on understanding the relationship between the airway microbiome and neutrophils.
Collapse
Affiliation(s)
- Guan-Zhen Xue
- School of Medicine, Qinghai University, Research Center for High Altitude Medicine, No.16 Kunlun Road, Xining, Qinghai Province, China
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining, Qinghai Province, China
| | - Hai-Zhen Ma
- Qinghai Provincial People's Hospital, Xining, Qinghai Province, China
| | - Ta-Na Wuren
- School of Medicine, Qinghai University, Research Center for High Altitude Medicine, No.16 Kunlun Road, Xining, Qinghai Province, China.
- Key Laboratory for Application for High Altitude Medicine, Qinghai University, Xining, Qinghai Province, China.
| |
Collapse
|
39
|
Malamud M, Whitehead L, McIntosh A, Colella F, Roelofs AJ, Kusakabe T, Dambuza IM, Phillips-Brookes A, Salazar F, Perez F, Shoesmith R, Zakrzewski P, Sey EA, Rodrigues C, Morvay PL, Redelinghuys P, Bedekovic T, Fernandes MJG, Almizraq R, Branch DR, Amulic B, Harvey J, Stewart D, Yuecel R, Reid DM, McConnachie A, Pickering MC, Botto M, Iliev ID, McInnes IB, De Bari C, Willment JA, Brown GD. Recognition and control of neutrophil extracellular trap formation by MICL. Nature 2024; 633:442-450. [PMID: 39143217 PMCID: PMC11390483 DOI: 10.1038/s41586-024-07820-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Regulation of neutrophil activation is critical for disease control. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA and neutrophil-derived proteins, are formed following pro-inflammatory signals; however, if this process is uncontrolled, NETs contribute to disease pathogenesis, exacerbating inflammation and host tissue damage1,2. Here we show that myeloid inhibitory C-type lectin-like (MICL), an inhibitory C-type lectin receptor, directly recognizes DNA in NETs; this interaction is vital to regulate neutrophil activation. Loss or inhibition of MICL functionality leads to uncontrolled NET formation through the ROS-PAD4 pathway and the development of an auto-inflammatory feedback loop. We show that in the context of rheumatoid arthritis, such dysregulation leads to exacerbated pathology in both mouse models and in human patients, where autoantibodies to MICL inhibit key functions of this receptor. Of note, we also detect similarly inhibitory anti-MICL autoantibodies in patients with other diseases linked to aberrant NET formation, including lupus and severe COVID-19. By contrast, dysregulation of NET release is protective during systemic infection with the fungal pathogen Aspergillus fumigatus. Together, we show that the recognition of NETs by MICL represents a fundamental autoregulatory pathway that controls neutrophil activity and NET formation.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/metabolism
- Aspergillus fumigatus/immunology
- Aspergillus fumigatus/pathogenicity
- Autoantibodies/immunology
- Autoantibodies/pharmacology
- COVID-19/immunology
- COVID-19/virology
- Disease Models, Animal
- DNA/metabolism
- DNA/immunology
- Extracellular Traps/metabolism
- Extracellular Traps/immunology
- Feedback, Physiological
- Inflammation/immunology
- Inflammation/metabolism
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/deficiency
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Mice, Inbred C57BL
- Neutrophil Activation
- Neutrophils/immunology
- Neutrophils/metabolism
- Protein-Arginine Deiminase Type 4/metabolism
- Reactive Oxygen Species/metabolism
- Receptors, Mitogen/antagonists & inhibitors
- Receptors, Mitogen/deficiency
- Receptors, Mitogen/immunology
- Receptors, Mitogen/metabolism
Collapse
Affiliation(s)
- Mariano Malamud
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Lauren Whitehead
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alasdair McIntosh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Fabio Colella
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Anke J Roelofs
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Takato Kusakabe
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease (JRI), Weill Cornell Medicine, New York City, NY, USA
| | - Ivy M Dambuza
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Fabián Salazar
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Federico Perez
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Romey Shoesmith
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Emily A Sey
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Petruta L Morvay
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Tina Bedekovic
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Maria J G Fernandes
- Faculty of Medicine, Department of Microbiology, Infectious Diseases, and Immunology, Laval University, Quebec City, Quebec, Canada
| | - Ruqayyah Almizraq
- Medical Affairs and Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Donald R Branch
- Medical Affairs and Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Borko Amulic
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Jamie Harvey
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Diane Stewart
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Raif Yuecel
- Centre for Cytomics, University of Exeter, Exeter, UK
| | - Delyth M Reid
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alex McConnachie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Matthew C Pickering
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Marina Botto
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease (JRI), Weill Cornell Medicine, New York City, NY, USA
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Cosimo De Bari
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Janet A Willment
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Gordon D Brown
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK.
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
40
|
Leal APF, Nieto Marín V, Cabistany VV, Morales J, Buccini DF, Franco OL. Applicability of mouse models for induction of severe acute lung injury. Pulm Pharmacol Ther 2024; 86:102316. [PMID: 39069252 DOI: 10.1016/j.pupt.2024.102316] [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: 04/17/2024] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Acute lung injury (ALI) is a significant clinical challenge associated with high morbidity and mortality. Worldwide, it affects approximately 200.000 individuals annually, with a staggering 40 % mortality rate in hospitalized cases and persistent complications in out-of-hospital cases. This review focuses on the key immunological pathways underlying bacterial ALI and the exploration of mouse models as tools for its induction. These models serve as indispensable platforms for unraveling the inflammatory cascades and biological responses inherent to ALI, while also facilitating the evaluation of novel therapeutic agents. However, their utility is not without challenges, mainly due to the stringent biosafety protocols required by the diverse bacterial virulence profiles. Simple and reproducible models of pulmonary bacterial infection are currently available, including intratracheal, intranasal, pleural and, intraperitoneal approaches. These models use endotoxins such as commercially available lipopolysaccharide (LPS) or live pathogens such as Pseudomonas aeruginosa, Mycobacterium tuberculosis, and Streptococcus pneumoniae, all of which are implicated in the pathogenesis of ALI. Combining murine models of bacterial lung infection with in-depth studies of the underlying immunological mechanisms is a cornerstone in advancing the therapeutic landscape for acute bacterial lung injury.
Collapse
Affiliation(s)
- Ana Paula Ferreira Leal
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil
| | - Valentina Nieto Marín
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil
| | - Vinícius Varzim Cabistany
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil
| | - Júlia Morales
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil
| | - Danieli Fernanda Buccini
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil
| | - Octávio Luiz Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Pontifícia Universidade Católica Dom Bosco, Campo Grande, MS, 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Pontifícia Universidade Católica de Brasília, Brasília, DF, 70790160, Brazil.
| |
Collapse
|
41
|
Sayson SG, Ashbaugh A, Porollo A, Smulian G, Cushion MT. Pneumocystis murina promotes inflammasome formation and NETosis during Pneumocystis pneumonia. mBio 2024; 15:e0140924. [PMID: 38953359 PMCID: PMC11323544 DOI: 10.1128/mbio.01409-24] [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: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Pneumocystis jirovecii pneumonia (PjP) poses a serious risk to individuals with compromised immune systems, such as individuals with HIV/AIDS or undergoing immunosuppressive therapies for cancer or solid organ transplants. Severe PjP triggers excessive lung inflammation, resulting in lung function decline and consequential alveolar damage, potentially culminating in acute respiratory distress syndrome. Non-HIV patients face a 30%-60% mortality rate, emphasizing the need for a deeper understanding of inflammatory responses in PjP. Prior research emphasized macrophages in Pneumocystis infections, neglecting neutrophils' role in tissue damage. Consequently, the overemphasis on macrophages led to an incomplete understanding of the role of neutrophils and inflammatory responses. In the current investigation, our RNAseq studies on a murine surrogate model of PjP revealed heightened activation of the NLRP3 inflammasome and NETosis cell death pathways in their lungs. Immunofluorescence staining confirmed neutrophil extracellular trap (NET) presence in the lungs of the P. murina-infected mice, validating our findings. Moreover, isolated neutrophils exhibited NETosis when directly stimulated with P. murina. Isolated NETs compromised P. murina viability in vitro, highlighting the potential role of neutrophils in controlling fungal growth and promoting inflammation during P. murina pneumonia through NLRP3 inflammasome assembly and NETosis. These pathways, essential for inflammation and pathogen elimination, bear the risk of uncontrolled activation leading to excessive tissue damage and persistent inflammation. This pioneering study is the first to identify the formation of NETs and inflammasomes during Pneumocystis infection, paving the way for comprehensive investigations into treatments aimed at mitigating lung damage and augmenting survival rates for individuals with PjP.IMPORTANCEPneumocystis jirovecii pneumonia (PjP) affects individuals with weakened immunity, such as HIV/AIDS, cancer, and organ transplant patients. Severe PjP triggers lung inflammation, impairing function and potentially causing acute respiratory distress syndrome. Non-HIV individuals face a 30%-60% mortality rate, underscoring the need for deeper insight into PjP's inflammatory responses. Past research focused on macrophages in managing Pneumocystis infection and its inflammation, while the role of neutrophils was generally overlooked. In contrast, our findings in P. murina-infected mouse lungs showed neutrophil involvement during inflammation and increased expression of NLRP3 inflammasome and NETosis pathways. Detection of neutrophil extracellular traps further indicated their involvement in the inflammatory process. Although beneficial in combating infection, unregulated neutrophil activation poses a potential threat to lung tissues. Understanding the behavior of neutrophils in Pneumocystis infections is crucial for controlling detrimental reactions and formulating treatments to reduce lung damage, ultimately improving the survival rates of individuals with PjP.
Collapse
Affiliation(s)
- Steven G. Sayson
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Alan Ashbaugh
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Aleksey Porollo
- Division of Human Genetics, Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - George Smulian
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Melanie T. Cushion
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- The Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Coupland LA, Spiro C, Quah BJC, Orlov A, Browne A, O'Meara CH, Kang CW, Frost S, Schulz L, Lombardo L, Parish CR, Aneman A. PLASMA DYNAMICS OF NEUTROPHIL EXTRACELLULAR TRAPS AND CELL-FREE DNA IN SEPTIC AND NONSEPTIC VASOPLEGIC SHOCK: A PROSPECTIVE COMPARATIVE OBSERVATIONAL COHORT STUDY. Shock 2024; 62:193-200. [PMID: 38813920 DOI: 10.1097/shk.0000000000002396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
ABSTRACT Background: The association between neutrophil extracellular traps (NETs) and the requirement for vasopressor and inotropic support in vasoplegic shock is unclear. This study aimed to investigate the dynamics of plasma levels of NETs and cell-free DNA (cfDNA) up to 48 h after the admission to the intensive care unit (ICU) for management of vasoplegic shock of infectious (SEPSIS) or noninfectious (following cardiac surgery, CARDIAC) origin. Methods: This is a prospective, observational study of NETs and cfDNA plasma levels at 0H (admission) and then at 12H, 24H, and 48H in SEPSIS and CARDIAC patients. The vasopressor inotropic score (VIS), the Sequential Organ Failure Assessment (SOFA) score, and time spent with invasive ventilation, in ICU and in hospital, were recorded. Associations between NETs/cfDNA and VIS and SOFA were analyzed by Spearman's correlation (rho), and between NETs/cfDNA and ventilation/ICU/hospitalization times by generalized linear regression. Results: Both NETs and cfDNA remained elevated over 48 h in SEPSIS (n = 46) and CARDIAC (n = 30) patients, with time-weighted average concentrations greatest in SEPSIS (NETs median difference 0.06 [0.02-0.11], P = 0.005; cfDNA median difference 0.48 [0.20-1.02], P < 0.001). The VIS correlated to NETs (rho = 0.3-0.60 in SEPSIS, P < 0.01, rho = 0.36-0.57 in CARDIAC, P ≤ 0.01) and cfDNA (rho = 0.40-0.56 in SEPSIS, P < 0.01, rho = 0.38-0.47 in CARDIAC, P < 0.05). NETs correlated with SOFA. Neither NETs nor cfDNA were independently associated with ventilator/ICU/hospitalization times. Conclusion: Plasma levels of NETs and cfDNA correlated with the dose of vasopressors and inotropes administered over 48 h in patients with vasoplegic shock from sepsis or following cardiac surgery. NETs levels also correlated with organ dysfunction. These findings suggest that similar mechanisms involving release of NETs are involved in the pathophysiology of vasoplegic shock irrespective of an infectious or noninfectious etiology.
Collapse
Affiliation(s)
| | - Calista Spiro
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Benjamin J-C Quah
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Anna Orlov
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Anna Browne
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Connor H O'Meara
- Department of Otorhinolaryngology, Head & Neck Surgery, The Canberra Hospital, Garran, ACT, Australia
| | - Chang-Won Kang
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | | | - Luis Schulz
- Intensive Care Unit, Liverpool Hospital, Sydney, Australia
| | - Lien Lombardo
- Intensive Care Unit, Liverpool Hospital, Sydney, Australia
| | - Christopher R Parish
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | | |
Collapse
|
43
|
Niri P, Saha A, Polopalli S, Kumar M, Das S, Chattopadhyay P. Role of biomarkers and molecular signaling pathways in acute lung injury. Fundam Clin Pharmacol 2024; 38:640-657. [PMID: 38279523 DOI: 10.1111/fcp.12987] [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: 04/03/2023] [Revised: 12/07/2023] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is caused by bacterial, fungal, and viral infections. When pathogens invade the lungs, the immune system responds by producing cytokines, chemokines, and interferons to promote the infiltration of phagocytic cells, which are essential for pathogen clearance. Their excess production causes an overactive immune response and a pathological hyper-inflammatory state, which leads to ALI. Until now, there is no particular pharmaceutical treatment available for ALI despite known inflammatory mediators like neutrophil extracellular traps (NETs) and reactive oxygen species (ROS). OBJECTIVES Therefore, the primary objective of this review is to provide the clear overview on the mechanisms controlling NETs, ROS formation, and other relevant processes during the pathogenesis of ALI. In addition, we have discussed the significance of epithelial and endothelial damage indicators and several molecular signaling pathways associated with ALI. METHODS The literature review was done from Web of Science, Scopus, PubMed, and Google Scholar for ALI, NETs, ROS, inflammation, biomarkers, Toll- and nucleotide-binding oligomerization domain (NOD)-like receptors, alveolar damage, pro-inflammatory cytokines, and epithelial/endothelial damage alone or in combination. RESULTS This review summarized the main clinical signs of ALI, including the regulation and distinct function of epithelial and endothelial biomarkers, NETs, ROS, and pattern recognition receptors (PRRs). CONCLUSION However, no particular drugs including vaccine for ALI has been established. Furthermore, there is a lack of validated diagnostic tools and a poor predictive rationality of current therapeutic biomarkers. Hence, extensive and precise research is required to speed up the process of drug testing and development by the application of artificial intelligence technologies, structure-based drug design, in-silico approaches, and drug repurposing.
Collapse
Affiliation(s)
- Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Subramanyam Polopalli
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, India
| | - Sanghita Das
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
| |
Collapse
|
44
|
Taenaka H, Fang X, Maishan M, Trivedi A, Wick KD, Gotts JE, Martin TR, Calfee CS, Matthay MA. Neutrophil reduction attenuates the severity of lung injury in the early phase of pneumococcal pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2024; 327:L141-L149. [PMID: 38772909 PMCID: PMC11687957 DOI: 10.1152/ajplung.00113.2024] [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: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
Neutrophils are the first leukocytes to be recruited to sites of inflammation in response to chemotactic factors released by activated macrophages and pulmonary epithelial and endothelial cells in bacterial pneumonia, a common cause of acute respiratory distress syndrome (ARDS). Although neutrophilic inflammation facilitates the elimination of pathogens, neutrophils also may cause bystander tissue injury. Even though the presence of neutrophils in alveolar spaces is a key feature of acute lung injury and ARDS especially from pneumonia, their contribution to the pathogenesis of lung injury is uncertain. The goal of this study was to elucidate the role of neutrophils in a clinically relevant model of bacterial pneumonia. We investigated the effect of reducing neutrophils in a mouse model of pneumococcal pneumonia treated with antibiotics. Neutrophils were reduced with anti-lymphocyte antigen 6 complex locus G6D (Ly6G) monoclonal antibody 24 h before and immediately preceding infection. Mice were inoculated intranasally with Streptococcus pneumoniae and received ceftriaxone 12 h after bacterial inoculation. Neutrophil reduction in mice treated with ceftriaxone attenuated hypoxemia, alveolar permeability, epithelial injury, pulmonary edema, and inflammatory biomarker release induced by bacterial pneumonia, even though bacterial loads in the distal air spaces of the lung were modestly increased as compared with antibiotic treatment alone. Thus, when appropriate antibiotics are administered, lung injury in the early phase of bacterial pneumonia is mediated in part by neutrophils. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.NEW & NOTEWORTHY Neutrophil accumulation is a key feature of ARDS, but their contribution to the pathogenesis is still uncertain. We investigated the effect of reducing neutrophils in a clinically relevant mouse model of pneumococcal pneumonia treated with antibiotics. When appropriate antibiotics were administered, neutrophil reduction with Ly6G antibody markedly attenuated lung injury and improved oxygenation. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Xiaohui Fang
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Mazharul Maishan
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Alpa Trivedi
- Department of Laboratory Medicine, University of California, San Francisco, California, United States
| | - Katherine D Wick
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Jeffrey E Gotts
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Thomas R Martin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Carolyn S Calfee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Michael A Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| |
Collapse
|
45
|
Thierry AR, Salmon D. Inflammation-, immunothrombosis,- and autoimmune-feedback loops may lead to persistent neutrophil self-stimulation in long COVID. J Med Virol 2024; 96:e29887. [PMID: 39189651 DOI: 10.1002/jmv.29887] [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: 03/28/2024] [Revised: 07/10/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Understanding the pathophysiology of long COVID is one of the most intriguing challenges confronting contemporary medicine. Despite observations recently made in the relevant molecular, cellular, and physiological domains, it is still difficult to say whether the post-acute sequelae of COVID-19 directly correspond to the consequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This work hypothesizes that neutrophils and neutrophil extracellular traps (NETs) production are at the interconnection of three positive feedback loops which are initiated in the acute phase of SARS-CoV-2 infection, and which involve inflammation, immunothrombosis, and autoimmunity. This phenomenon could be favored by the fact that SARS-CoV-2 may directly bind and penetrate neutrophils. The ensuing strong neutrophil stimulation leads to a progressive amplification of an exacerbated and uncontrolled NETs production, potentially persisting for months beyond the acute phase of infection. This continuous self-stimulation of neutrophils leads, in turn, to systemic inflammation, micro-thromboses, and the production of autoantibodies, whose significant consequences include the persistence of endothelial and multiorgan damage, and vascular complications.
Collapse
Affiliation(s)
- Alain R Thierry
- IRCM, Institute of Research on Cancerology of Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
- Montpellier Cancer Institute (ICM), Montpellier, France
| | | |
Collapse
|
46
|
Turner DL, Amoozadeh S, Baric H, Stanley E, Werder RB. Building a human lung from pluripotent stem cells to model respiratory viral infections. Respir Res 2024; 25:277. [PMID: 39010108 PMCID: PMC11251358 DOI: 10.1186/s12931-024-02912-0] [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: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.
Collapse
Affiliation(s)
- Declan L Turner
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Sahel Amoozadeh
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Hannah Baric
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Ed Stanley
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, 3056, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia.
| |
Collapse
|
47
|
Li Q, Gan Q, Chi H, Meng X, Dalmo RA, Sheng X, Tang X, Xing J, Zhan W. Extracellular traps in skin lesions infected with lymphocystis disease virus in black rockfish (Sebastes schlegelii). FISH & SHELLFISH IMMUNOLOGY 2024; 150:109643. [PMID: 38763177 DOI: 10.1016/j.fsi.2024.109643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
The lymphocystis disease (LCD), caused by Lymphocystis disease virus (LCDV), is a benign and self-limiting disease described in a many freshwater and marine fish species. Hypertrophic fibroblasts and extensive aggregation of inflammatory cells are characteristics of LCD. In the present study, small animal imaging and ultrastructural investigations were carried out on the lymphocystis nodules of black rockfish (Sebastes schlegelii) naturally infected with lymphocystis iridovirus, to assess pathology, and the exudate with particular attention to the formation of extracellular traps (ETs) in vivo. Ex vivo were examined by nodules sections and primary cells stimulation. By histopathological analysis, the nodules contained infiltrated inflammatory cells and extensive basophilic fibrillar filaments at the periphery of the hypertrophied fibroblasts. ETs were assessed in nodules samples using indirect immunofluorescence to detect DNA and myeloperoxidase. Moreover, LCDV was able to infect peritoneal cells of black rockfish in vitro and induce the formation of ETs within 4 h. In summary, this study proved that ETs are involved in the response to LCDV infection and may be involved in formation of lymphoid nodules. Taken together, the findings provide a new perspective to determine the impact factors on the growth of nodules.
Collapse
Affiliation(s)
- Qian Li
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Qiujie Gan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Xianghu Meng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China
| | - Roy Ambli Dalmo
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, University of Tromsø - The Artic University of Norway, Tromsø, Norway
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
48
|
Toya S, Struyf S, Huerta L, Morris P, Gavioli E, Minnella EM, Cesta MC, Allegretti M, Proost P. A narrative review of chemokine receptors CXCR1 and CXCR2 and their role in acute respiratory distress syndrome. Eur Respir Rev 2024; 33:230172. [PMID: 39048127 PMCID: PMC11267298 DOI: 10.1183/16000617.0172-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe form of acute respiratory failure characterised by extensive inflammatory injury to the alveolocapillary barrier leading to alveolar oedema, impaired gas exchange and, ultimately, hypoxaemia necessitating the use of supplemental oxygen combined with some degree of positive airway pressure. Although much heterogeneity exists regarding the aetiology, localisation and endotypic characterisation of ARDS, what remains largely undisputed is the role of the innate immune system, and in particular of neutrophils, in precipitating and propagating lung injury. Activated neutrophils, recruited to the lung through chemokine gradients, promote injury by releasing oxidants, proteases and neutrophil extracellular traps, which ultimately cause platelet aggregation, microvascular thrombosis and cellular death. Among various neutrophilic chemoattractants, interleukin-8/C-X-C motif ligand 8 and related chemokines, collectively called ELR+ chemokines, acting on neutrophils through the G protein-coupled receptors CXCR1 and CXCR2, are pivotal in orchestrating the neutrophil activation status and chemotaxis in the inflamed lung. This allows efficient elimination of infectious agents while at the same time minimising collateral damage to host tissue. Therefore, understanding how CXCR1 and CXCR2 receptors are regulated is important if we hope to effectively target them for therapeutic use in ARDS. In the following narrative review, we provide an overview of the role of ELR+ chemokines in acute lung injury (ALI) and ARDS, we summarise the relevant regulatory pathways of their cognisant receptors CXCR1/2 and highlight current preclinical and clinical evidence on the therapeutic role of CXCR1 and CXCR2 inhibition in animal models of ALI, as well as in ARDS patients.
Collapse
Affiliation(s)
| | - Sofie Struyf
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Luis Huerta
- Keck School of Medicine of USC, Department of Medicine, Pulmonary and Critical Care Medicine, Los Angeles, CA, USA
| | - Peter Morris
- The University of Alabama at Birmingham, Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, Birmingham, AL, USA
| | | | | | | | | | - Paul Proost
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| |
Collapse
|
49
|
Chu JY, McCormick B, Sundaram K, Hardisty G, Karmakar U, Pumpe C, Krull E, Lucas CD, Amado-Azevedo J, Hordijk PL, Caporali A, Mellor H, Baillie JK, Rossi AG, Vermeren S. ARAP3 protects from excessive formylated peptide-induced microvascular leakage by acting on endothelial cells and neutrophils. J Pathol 2024; 263:347-359. [PMID: 38734878 DOI: 10.1002/path.6288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/31/2024] [Accepted: 03/25/2024] [Indexed: 05/13/2024]
Abstract
Vascular permeability is temporarily heightened during inflammation, but excessive inflammation-associated microvascular leakage can be detrimental, as evidenced in the inflamed lung. Formylated peptides regulate vascular leakage indirectly via formylated peptide receptor-1 (FPR1)-mediated recruitment and activation of neutrophils. Here we identify how the GTPase-activating protein ARAP3 protects against formylated peptide-induced microvascular permeability via endothelial cells and neutrophils. In vitro, Arap3-/- endothelial monolayers were characterised by enhanced formylated peptide-induced permeability due to upregulated endothelial FPR1 and enhanced vascular endothelial cadherin internalisation. In vivo, enhanced inflammation-associated microvascular leakage was observed in Arap3-/- mice. Leakage of plasma protein into the lungs of Arap3-/- mice increased within hours of formylated peptide administration. Adoptive transfer experiments indicated this was dependent upon ARAP3 deficiency in both immune and non-immune cells. Bronchoalveolar lavages of formylated peptide-challenged Arap3-/- mice contained neutrophil extracellular traps (NETs). Pharmacological inhibition of NET formation abrogated excessive microvascular leakage, indicating a critical function of NETs in this context. The observation that Arap3-/- mice developed more severe influenza suggests these findings are pertinent to pathological situations characterised by abundant formylated peptides. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Julia Y Chu
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Barry McCormick
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Kruthika Sundaram
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Gareth Hardisty
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Utsa Karmakar
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Caroline Pumpe
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Krull
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Christopher D Lucas
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Joana Amado-Azevedo
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Peter L Hordijk
- Department of Physiology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Andrea Caporali
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Harry Mellor
- School of Biochemistry, University of Bristol, Bristol, UK
| | - J Kenneth Baillie
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Sonja Vermeren
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
50
|
Huang L, Tan X, Xuan W, Luo Q, Xie L, Xi Y, Li R, Li L, Li F, Zhao M, Jiang Y, Wu X. Ficolin-A/2 Aggravates Severe Lung Injury through Neutrophil Extracellular Traps Mediated by Gasdermin D-Induced Pyroptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:989-1006. [PMID: 38442803 DOI: 10.1016/j.ajpath.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Neutrophil extracellular traps (NETs) and pyroptosis are critical events in lung injury. This study investigated whether ficolin-A influenced NET formation through pyroptosis to exacerbate lipopolysaccharide (LPS)-induced lung injury. The expression of ficolin-A/2, NETs, and pyroptosis-related molecules was investigated in animal and cell models. Knockout and knockdown (recombinant protein) methods were used to elucidate regulatory mechanisms. The Pearson correlation coefficient was used to analyze the correlation between ficolins and pyroptosis- and NET-related markers in clinical samples. In this study, ficolin-2 (similar to ficolin-A) showed significant overexpression in patients with acute respiratory distress syndrome. In vivo, knockout of Fcna, but not Fcnb, attenuated lung inflammation and inhibited NET formation in the LPS-induced mouse model. DNase I further alleviated lung inflammation and NET formation in Fcna knockout mice. In vitro, neutrophils derived from Fcna-/- mice showed less pyroptosis and necroptosis than those from the control group after LPS stimulation. Additionally, GSDMD knockdown or Nod-like receptor protein 3 inhibitor reduced NET formation. Addition of recombinant ficolin-2 protein to human peripheral blood neutrophils promoted NET formation and pyroptosis after LPS stimulation, whereas Fcn2 knockdown had the opposite effect. Acute respiratory distress syndrome patients showed increased levels of pyroptosis- and NET-related markers, which were correlated positively with ficolin-2 levels. In conclusion, these results suggested that ficolin-A/2 exacerbated NET formation and LPS-induced lung injury via gasdermin D-mediated pyroptosis.
Collapse
Affiliation(s)
- Li Huang
- Department of Pediatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xiaowu Tan
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Weixia Xuan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Luo
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Xie
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yunzhu Xi
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Rong Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Feifan Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Meiyun Zhao
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yongliang Jiang
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.
| | - Xu Wu
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|