51
|
Zheng J, Li Y, Kong X, Guo J. Exploring immune-related pathogenesis in lung injury: Providing new insights Into ALI/ARDS. Biomed Pharmacother 2024; 175:116773. [PMID: 38776679 DOI: 10.1016/j.biopha.2024.116773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) represent a significant global burden of morbidity and mortality, with lung injury being the primary cause of death in affected patients. The pathogenesis of lung injury, however, remains a complex issue. In recent years, the role of the immune system in lung injury has attracted extensive attention worldwide. Despite advancements in our understanding of various lung injury subtypes, significant limitations persist in both prevention and treatment. This review investigates the immunopathogenesis of ALI/ARDS, aiming to elucidate the pathological processes of lung injury mediated by dendritic cells (DCs), natural killer (NK) cells, phagocytes, and neutrophils. Furthermore, the article expounds on the critical contributions of gut microbiota, inflammatory pathways, and cytokine storms in the development of ALI/ARDS.
Collapse
Affiliation(s)
- Jiajing Zheng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Li
- Pharmacy Department of the First Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China
| | - Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jinhe Guo
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
52
|
Moriyama E, Nadatani Y, Higashimori A, Otani K, Ominami M, Fukunaga S, Hosomi S, Tanaka F, Taira K, Fujiwara Y, Watanabe T. Neutrophil extracellular trap formation and its implications in nonsteroidal anti-inflammatory drug-induced small intestinal injury. J Gastroenterol Hepatol 2024; 39:1123-1133. [PMID: 38576269 DOI: 10.1111/jgh.16543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/01/2024] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND AND AIM Nonsteroidal anti-inflammatory drugs (NSAIDs) damage the small intestine via neutrophil infiltration driven by the mucosal invasion of enterobacteria. The antimicrobial function of neutrophils is partially dependent on neutrophil extracellular traps (NETs). Excessive NET formation has been associated with several inflammatory diseases. Here, we aimed to investigate the role of NETs in NSAID-induced small intestinal damage using human samples and an experimental mouse model. METHODS Human small intestine specimens were obtained from NSAID users during double-balloon enteroscopy. Wild-type, protein arginine deiminase 4 (PAD4) knockout, and antibiotic-treated mice were administered indomethacin to induce small intestinal injury. The expression of NET-associated proteins, including PAD4, citrullinated histone H3 (CitH3), cell-free DNA, and myeloperoxidase (MPO), was evaluated. RESULTS The double-positive stained area with CitH3 and MPO, which is specific for neutrophil-derived extracellular traps, was significantly high in the injured small intestinal mucosa of NSAID users. In a mouse model, small intestinal damage developed at 6 h after indomethacin administration, accompanied by increased mRNA levels of interleukin-1β and keratinocyte chemoattractant and elevated NET-associated protein levels of PAD4, CitH3, and MPO in small intestine and serum levels of cell-free DNA. Both genetic deletion and pharmacological inhibition of PAD4 attenuated this damage by reducing the mRNA expression of inflammatory cytokines and NET-associated proteins. Furthermore, mice pretreated with antibiotics showed resistance to indomethacin-induced small intestinal damage, with less NET formation. CONCLUSION These results suggest that NETs aggravate NSAID-induced small intestinal injury. Therefore, NET inhibition could be a potential treatment for NSAID-induced small intestinal injury.
Collapse
Affiliation(s)
- Eiji Moriyama
- Department of Gastroenterology, Graduate School of Medicine, Osaka city University, Osaka, Japan
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Higashimori
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Koji Otani
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Masaki Ominami
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shusei Fukunaga
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Koichi Taira
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Toshio Watanabe
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
53
|
Moussavi-Harami SF, Cleary SJ, Magnen M, Seo Y, Conrad C, English BC, Qiu L, Wang KM, Abram CL, Lowell CA, Looney MR. Loss of neutrophil Shp1 produces hemorrhagic and lethal acute lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.595575. [PMID: 38854059 PMCID: PMC11160570 DOI: 10.1101/2024.05.23.595575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The 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 knockout 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 F Moussavi-Harami
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of California, San Francisco
| | - S J Cleary
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - M Magnen
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - Y Seo
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - C Conrad
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - B C English
- Department of Microbiology & Immunology, University of California, San Francisco
- CoLabs, University of California, San Francisco
| | - L Qiu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - K M Wang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
| | - C L Abram
- Department of Laboratory Medicine, University of California, San Francisco
| | - C A Lowell
- Department of Laboratory Medicine, University of California, San Francisco
| | - M R Looney
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco
- Department of Laboratory Medicine, University of California, San Francisco
| |
Collapse
|
54
|
Qiao X, Yin J, Zheng Z, Li L, Feng X. Endothelial cell dynamics in sepsis-induced acute lung injury and acute respiratory distress syndrome: pathogenesis and therapeutic implications. Cell Commun Signal 2024; 22:241. [PMID: 38664775 PMCID: PMC11046830 DOI: 10.1186/s12964-024-01620-y] [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: 02/28/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis, a prevalent critical condition in clinics, continues to be the leading cause of death from infections and a global healthcare issue. Among the organs susceptible to the harmful effects of sepsis, the lungs are notably the most frequently affected. Consequently, patients with sepsis are predisposed to developing acute lung injury (ALI), and in severe cases, acute respiratory distress syndrome (ARDS). Nevertheless, the precise mechanisms associated with the onset of ALI/ARDS remain elusive. In recent years, there has been a growing emphasis on the role of endothelial cells (ECs), a cell type integral to lung barrier function, and their interactions with various stromal cells in sepsis-induced ALI/ARDS. In this comprehensive review, we summarize the involvement of endothelial cells and their intricate interplay with immune cells and stromal cells, including pulmonary epithelial cells and fibroblasts, in the pathogenesis of sepsis-induced ALI/ARDS, with particular emphasis placed on discussing the several pivotal pathways implicated in this process. Furthermore, we discuss the potential therapeutic interventions for modulating the functions of endothelial cells, their interactions with immune cells and stromal cells, and relevant pathways associated with ALI/ARDS to present a potential therapeutic strategy for managing sepsis and sepsis-induced ALI/ARDS.
Collapse
Affiliation(s)
- Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Liangge Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
55
|
King PT, Dousha L. Neutrophil Extracellular Traps and Respiratory Disease. J Clin Med 2024; 13:2390. [PMID: 38673662 PMCID: PMC11051312 DOI: 10.3390/jcm13082390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/26/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Extracellular traps made by neutrophils (NETs) and other leukocytes such as macrophages and eosinophils have a key role in the initial immune response to infection but are highly inflammatory and may contribute to tissue damage. They are particularly relevant to lung disease, with the pulmonary anatomy facilitating their ability to fully extend into the airways/alveolar space. There has been a rapid expansion in the number of published studies demonstrating their role in a variety of important respiratory diseases including chronic obstructive pulmonary disease, cystic fibrosis, bronchiectasis, asthma, pneumonia, COVID-19, rhinosinusitis, interstitial lung disease and lung cancer. The expression of NETs and other traps is a specific process, and diagnostic tests need to differentiate them from other inflammatory pathways/causes of cell death that are also characterised by the presence of extracellular DNA. The specific targeting of this pathway by relevant therapeutics may have significant clinical benefit; however, current clinical trials/evidence are at a very early stage. This review will provide a broad overview of the role of NETs and their possible treatment in respiratory disease.
Collapse
Affiliation(s)
- Paul T. King
- Monash Lung, Sleep, Allergy and Immunology, Monash Medical Centre, 246 Clayton Rd, Clayton, Melbourne, VIC 3168, Australia;
- Department of Medicine, Monash University, Clayton, Melbourne, VIC 3168, Australia
| | - Lovisa Dousha
- Monash Lung, Sleep, Allergy and Immunology, Monash Medical Centre, 246 Clayton Rd, Clayton, Melbourne, VIC 3168, Australia;
- Department of Medicine, Monash University, Clayton, Melbourne, VIC 3168, Australia
| |
Collapse
|
56
|
Alexa AL, Sargarovschi S, Ionescu D. Neutrophils and Anesthetic Drugs: Implications in Onco-Anesthesia. Int J Mol Sci 2024; 25:4033. [PMID: 38612841 PMCID: PMC11012681 DOI: 10.3390/ijms25074033] [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: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Apart from being a significant line of defense in the host defense system, neutrophils have many immunological functions. Although there are not many publications that accurately present the functions of neutrophils in relation to oncological pathology, their activity and implications have been studied a lot recently. This review aims to extensively describe neutrophils functions'; their clinical implications, especially in tumor pathology; the value of clinical markers related to neutrophils; and the implications of neutrophils in onco-anesthesia. This review also aims to describe current evidence on the influence of anesthetic drugs on neutrophils' functions and their potential influence on perioperative outcomes.
Collapse
Affiliation(s)
- Alexandru Leonard Alexa
- Department of Anesthesia and Intensive Care I, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (S.S.); (D.I.)
- Association for Research in Anesthesia and Intensive Care (ACATI), 400162 Cluj-Napoca, Romania
- Onco-Anaesthesia Research Group, ESAIC, 1000 Brussels, Belgium
| | - Sergiu Sargarovschi
- Department of Anesthesia and Intensive Care I, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (S.S.); (D.I.)
- Association for Research in Anesthesia and Intensive Care (ACATI), 400162 Cluj-Napoca, Romania
| | - Daniela Ionescu
- Department of Anesthesia and Intensive Care I, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (S.S.); (D.I.)
- Association for Research in Anesthesia and Intensive Care (ACATI), 400162 Cluj-Napoca, Romania
- Onco-Anaesthesia Research Group, ESAIC, 1000 Brussels, Belgium
- Outcome Research Consortium, Cleveland, OH 44195, USA
| |
Collapse
|
57
|
Wei S, Shen Z, Yin Y, Cong Z, Zeng Z, Zhu X. Advances of presepsin in sepsis-associated ARDS. Postgrad Med J 2024; 100:209-218. [PMID: 38147883 DOI: 10.1093/postmj/qgad132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/30/2023] [Accepted: 12/02/2023] [Indexed: 12/28/2023]
Abstract
This article reviews the correlation between presepsin and sepsis and the resulting acute respiratory distress syndrome (ARDS). ARDS is a severe complication of sepsis. Despite the successful application of protective mechanical ventilation, restrictive fluid therapy, and neuromuscular blockade, which have effectively reduced the morbidity and mortality associated with ARDS, the mortality rate among patients with sepsis-associated ARDS remains notably high. The challenge lies in the prediction of ARDS onset and the timely implementation of intervention strategies. Recent studies have demonstrated significant variations in presepsin (PSEP) levels between patients with sepsis and those without, particularly in the context of ARDS. Moreover, these studies have revealed substantially elevated PSEP levels in patients with sepsis-associated ARDS compared to those with nonsepsis-associated ARDS. Consequently, PSEP emerges as a valuable biomarker for identifying patients with an increased risk of sepsis-associated ARDS and to predict in-hospital mortality.
Collapse
Affiliation(s)
- Senhao Wei
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Graduate School of Peking University Health Science Center, Peking University Health Science Center, Beijing 100191, China
| | - Ziyuan Shen
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Graduate School of Peking University Health Science Center, Peking University Health Science Center, Beijing 100191, China
| | - Yiyuan Yin
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Zhukai Cong
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Zhaojin Zeng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Graduate School of Peking University Health Science Center, Peking University Health Science Center, Beijing 100191, China
| | - Xi Zhu
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
58
|
Busch MH, Ysermans R, Aendekerk JP, Timmermans SAMEG, Potjewijd J, Damoiseaux JGMC, Spronk HMH, ten Cate H, Reutelingsperger CP, Nagy M, van Paassen P. The intrinsic coagulation pathway plays a dominant role in driving hypercoagulability in ANCA-associated vasculitis. Blood Adv 2024; 8:1295-1304. [PMID: 38175623 PMCID: PMC10918483 DOI: 10.1182/bloodadvances.2023011937] [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: 10/17/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
ABSTRACT The risk of a venous thrombotic event (VTE) is increased in patients with antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV); however, a detailed understanding of the underlying mechanisms of hypercoagulability is limited. We assessed prospectively different coagulation parameters in 71 patients with active AAV at baseline and after 6 months of follow-up. D-dimers and fibrinogen were increased in most patients at presentation and remained elevated in half of the patients. Particularly, thrombin-antithrombin (T:AT) complex and activated coagulation factors in complex with their natural inhibitors of the intrinsic coagulation pathway (ie, activated FXII:C1 esterase inhibitor [FXIIa:C1Inh], FXIa:AT, and FXIa:alpha1-antitrypsin [FXIa:α1AT]) were profoundly elevated in patients at baseline. Thrombin formation was dominantly correlated with coagulation factors of the intrinsic pathway (ie, FXIIa:AT, FXIa:AT, FXIa:α1AT, and FXIa:C1Inh) compared to the extrinsic pathway (ie, FVIIa:AT). Hypercoagulability correlated with higher disease activity, ANCA levels, C-reactive protein, serum creatinine, and proteinuria. VTEs were observed in 5 out of 71 (7%) patients within 1 month (interquartile range, 1-5) after inclusion. Baseline T:AT levels were significantly higher in patients with VTE than in those without VTE (P = .044), but other clinical or laboratory markers were comparable between both groups. Hypercoagulability is dominantly characterized by activation of the intrinsic coagulation pathway and elevated D-dimers in active AAV. The driving factors of hypercoagulability are yet to be studied but are most likely related to an interplay of increased disease activity, vascular inflammation, and endothelial damage. Future targets for intervention could include inhibitors of the intrinsic coagulation pathway and compounds specifically reducing the hyperinflammatory state.
Collapse
Affiliation(s)
- Matthias H. Busch
- Department Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Renée Ysermans
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Joop P. Aendekerk
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Sjoerd A. M. E. G. Timmermans
- Department Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Judith Potjewijd
- Department Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan G. M. C. Damoiseaux
- Department of Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Henri M. H. Spronk
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Hugo ten Cate
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
- Department of Internal Medicine, Thrombosis Expertise Center, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Magdolna Nagy
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Pieter van Paassen
- Department Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| |
Collapse
|
59
|
Chen Z, Xiao G, Ao J. Resveratrol Attenuates Rheumatoid Arthritis Induce Neutrophil Extracellular Traps via TLR-4 Mediated Inflammation in C57BL/6 Mice. Physiol Res 2024; 73:91-104. [PMID: 38466008 PMCID: PMC11019621 DOI: 10.33549/physiolres.935172] [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: 06/23/2023] [Accepted: 10/17/2023] [Indexed: 04/26/2024] Open
Abstract
The objective of this study was to evaluate whether RSV inhibits neutrophil extracellular traps (NETs) that induce joint hyperalgesia in C57BL/6 mice after adjuvant-induced arthritis. A subplantar injection of Freund's complete adjuvant was administered to C57BL/6 mice on day 0 for immunization in the AIA model. Resveratrol (RSV, 25 mg/kg) was administered intraperitoneally once daily starting on day 22 and continuing for two weeks. The effects of mechanical hyperalgesia and edema formation have been assessed in addition to histopathological scoring. Mice were sacrificed on day 35 to determine cytokine levels and PADI4 and COX-2 expression levels. ELISA was used to quantify neutrophil extracellular traps (NETs) along with neutrophil elastase-DNA and myeloperoxidase-DNA complexes in neutrophils. An immunohistochemical stain was performed on knee joints to determine the presence of nuclear factor kappa B p65 (NF-kappaB p65). AIA mice were found to have higher levels of NET in joints and their joint cells demonstrated an increased expression of the PADI4 gene. Treatment with RSV in AIA mice (25 mg/kg, i.p.) significantly (P<0.05) inhibited joint hyperalgesia, resulting in a significant increase in mechanical threshold, a decrease in articular edema, a decrease in the production of inflammatory cytokines, increased COX-2 expression, and a decrease in the immunostaining of NF-kappaB. Furthermore, treatment with RSV significantly reduced the amount of neutrophil elastase (NE)-DNA and MPO-DNA complexes, which were used as indicators of NET formation (P<0.05). This study indicates that RSV reduces NET production and hyperalgesia by reducing inflammation mediated by PADI4 and COX-2. According to these data, NETs contribute to joint pain and resveratrol can be used to treat pain in RA through this pathway.
Collapse
Affiliation(s)
- Z Chen
- Department of Orthopedics, Wushan County Hospital of TCM, Chongqing, China.
| | | | | |
Collapse
|
60
|
Zheng Y, Li Y, Li M, Wang R, Jiang Y, Zhao M, Lu J, Li R, Li X, Shi S. COVID-19 cooling: Nanostrategies targeting cytokine storm for controlling severe and critical symptoms. Med Res Rev 2024; 44:738-811. [PMID: 37990647 DOI: 10.1002/med.21997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants continue to wreak havoc worldwide, the "Cytokine Storm" (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long-term effects of vaccines and broad-spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID-19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID-19 caused by CS.
Collapse
Affiliation(s)
- Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mao Li
- Health Management Centre, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
61
|
Rizo-Téllez SA, Filep JG. Beyond host defense and tissue injury: the emerging role of neutrophils in tissue repair. Am J Physiol Cell Physiol 2024; 326:C661-C683. [PMID: 38189129 PMCID: PMC11193466 DOI: 10.1152/ajpcell.00652.2023] [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: 11/29/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Neutrophils, the most abundant immune cells in human blood, play a fundamental role in host defense against invading pathogens and tissue injury. Neutrophils carry potentially lethal weaponry to the affected site. Inadvertent and perpetual neutrophil activation could lead to nonresolving inflammation and tissue damage, a unifying mechanism of many common diseases. The prevailing view emphasizes the dichotomy of their function, host defense versus tissue damage. However, tissue injury may also persist during neutropenia, which is associated with disease severity and poor outcome. Numerous studies highlight neutrophil phenotypic heterogeneity and functional versatility, indicating that neutrophils play more complex roles than previously thought. Emerging evidence indicates that neutrophils actively orchestrate resolution of inflammation and tissue repair and facilitate return to homeostasis. Thus, neutrophils mobilize multiple mechanisms to limit the inflammatory reaction, assure debris removal, matrix remodeling, cytokine scavenging, macrophage reprogramming, and angiogenesis. In this review, we will summarize the homeostatic and tissue-reparative functions and mechanisms of neutrophils across organs. We will also discuss how the healing power of neutrophils might be harnessed to develop novel resolution and repair-promoting therapies while maintaining their defense functions.
Collapse
Affiliation(s)
- Salma A Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| |
Collapse
|
62
|
Pan T, Lee JW. A crucial role of neutrophil extracellular traps in pulmonary infectious diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:34-41. [PMID: 39170960 PMCID: PMC11332830 DOI: 10.1016/j.pccm.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 08/23/2024]
Abstract
Neutrophil extracellular traps (NETs), extrusions of intracellular DNA with attached granular material that exert an antibacterial effect through entangling, isolating, and immobilizing microorganisms, have been extensively studied in recent decades. The primary role of NETs is to entrap and facilitate the killing of bacteria, fungi, viruses, and parasites, preventing bacterial and fungal dissemination. NET formation has been described in many pulmonary diseases, including both infectious and non-infectious. NETs are considered a double-edged sword. As innate immune cells, neutrophils release NETs to kill pathogens and remove cellular debris. However, the deleterious effects of excessive NET release in lung disease are particularly important because NETs and by-products of NETosis can directly induce epithelial and endothelial cell death while simultaneously inducing inflammatory cytokine secretion and immune-mediated thrombosis. Thus, NET formation must be tightly regulated to preserve the anti-microbial capability of NETs while minimizing damage to the host. In this review, we summarized the recent updates on the mechanism of NETs formation and pathophysiology associated with excessive NETs, aiming to provide insights for research and treatment of pulmonary infectious diseases.
Collapse
Affiliation(s)
- Ting Pan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jae Woo Lee
- Department of Anesthesiology, University of California Los Angeles, Los Angeles, CA 90230, USA
| |
Collapse
|
63
|
Haus M, Foltan M, Philipp A, Mueller T, Gruber M, Lingel MP, Krenkel L, Lehle K. Neutrophil extracellular traps - a potential trigger for the development of thrombocytopenia during extracorporeal membrane oxygenation. Front Immunol 2024; 15:1339235. [PMID: 38449869 PMCID: PMC10914994 DOI: 10.3389/fimmu.2024.1339235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Neutrophil extracellular traps (NETs) have recently emerged as a potential link between inflammation, immunity, and thrombosis, as well as other coagulation disorders which present a major challenge in the context of extracorporeal membrane oxygenation (ECMO). By examining blood from ECMO patients for NETs and their precursors and correlating them with clinical and laboratory biomarkers of coagulation and inflammation, this study aims to evaluate the association between the presence of NETs in the bloodstream of ECMO patients and the development of potentially severe coagulation disorders during ECMO therapy. Therefore, blood samples were collected from healthy volunteers (n=13) and patients receiving veno-venous (VV) ECMO therapy (n=10). To identify NETs and their precursors, DNA and myeloperoxidase as well as granulocyte marker CD66b were visualized simultaneously by immunofluorescence staining in serial blood smears. Differentiation of DNA-containing objects and identification of NETs and their precursors was performed semiautomatically by a specific algorithm using the shape and size of DNA staining and the intensity of MPO and CD66b signal. Neutrophil extracellular traps and their precursors could be detected in blood smears from patients requiring VV ECMO. Compared to volunteers, ECMO patients presented significantly higher rates of NETs and NET precursors as well as an increased proportion of neutrophil granulocytes in all detected nucleated cells. A high NET rate prior to the initiation of ECMO therapy was associated with both increased IL-6 and TNF-α levels as an expression of a high cytokine burden. These patients with increased NET release also presented an earlier and significantly more pronounced decrease in platelet counts and ATIII activity following initiation of therapy compared with patients with less elevated NETs. These findings provide further indications for the development of immune-mediated acquired thrombocytopenia in ECMO patients.
Collapse
Affiliation(s)
- Moritz Haus
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maik Foltan
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Alois Philipp
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Mueller
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Maximilian P. Lingel
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lars Krenkel
- Regensburg Center of Biomedical Engineering, Ostbayerische Technische Hochschule, Regensburg, Germany
| | - Karla Lehle
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
64
|
Ke D, Ni J, Yuan Y, Cao M, Chen S, Zhou H. Identification and Validation of Hub Genes Related to Neutrophil Extracellular Traps-Mediated Cell Damage During Myocardial Infarction. J Inflamm Res 2024; 17:617-637. [PMID: 38323113 PMCID: PMC10844013 DOI: 10.2147/jir.s444975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024] Open
Abstract
Purpose Studies have shown that neutrophil-mediated formation of neutrophil extracellular traps (NETs) leads to increased inflammatory response and cellular tissue damage during myocardial infarction (MI). We aimed to identify and validate possible hub genes in the process of NETs-mediated cell damage. Methods We performed an immune cell infiltration analysis of the MI transcriptome dataset based on CIBERSORT and ssGSEA algorithms. Gene expression profiles of NETs formation (GSE178883) were used to analyze the physiological processes of peripheral blood neutrophils after phorbol myristate acetate (PMA) stimulation. Bioinformatics and machine learning algorithms were utilized to find candidate hub genes based on NETs-related genes and transcriptome datasets (GSE66360 and GSE179828). We generated the receiver operating curve (ROC) to evaluate the diagnostic value of hub genes. Next, the correlation between hub genes and immune cells was analyzed using CIBERSORT, ssGSEA and xCell algorithms. Finally, we used quantitative real-time PCR (qRT-PCR) and immunohistochemistry to verify gene expression. Results Immune cell infiltration analysis revealed that inflammatory cells such as neutrophils were highly expressed in the peripheral blood of patients with MI. Functional analysis of differentially expressed genes (DEGs) in GSE178883 indicated that the potential pathogenesis lies in immune terms. Using weighted gene co-expression network analysis (WGCNA) and machine learning algorithms, we finally identified the seven hub genes (FCAR, IL1B, MMP9, NFIL3, CXCL2, ICAM1, and ZFP36). The qRT-PCR results showed that IL-1B, MMP9, and NFIL3 mRNA expression was up-regulated in the MI group compared to the control. Immunohistochemical results showed high MMP9, IL-1B, and NFIL3 expression in the infarcted area compared to the non-infarcted area and sham-operated groups. Conclusion We identified seven hub genes associated with NETs-mediated cellular damage during MI. Our results may provide insights into the mechanisms of neutrophil-mediated cell injury during MI.
Collapse
Affiliation(s)
- Da Ke
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Jian Ni
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Mingzhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Si Chen
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| |
Collapse
|
65
|
Tu H, Ren H, Jiang J, Shao C, Shi Y, Li P. Dying to Defend: Neutrophil Death Pathways and their Implications in Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306457. [PMID: 38044275 PMCID: PMC10885667 DOI: 10.1002/advs.202306457] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/06/2023] [Indexed: 12/05/2023]
Abstract
Neutrophils, accounting for ≈70% of human peripheral leukocytes, are key cells countering bacterial and fungal infections. Neutrophil homeostasis involves a balance between cell maturation, migration, aging, and eventual death. Neutrophils undergo different death pathways depending on their interactions with microbes and external environmental cues. Neutrophil death has significant physiological implications and leads to distinct immunological outcomes. This review discusses the multifarious neutrophil death pathways, including apoptosis, NETosis, pyroptosis, necroptosis, and ferroptosis, and outlines their effects on immune responses and disease progression. Understanding the multifaceted aspects of neutrophil death, the intersections among signaling pathways and ramifications of immunity will help facilitate the development of novel therapeutic methods.
Collapse
Affiliation(s)
- Haiyue Tu
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Haoyu Ren
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Junjie Jiang
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Peishan Li
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| |
Collapse
|
66
|
Zhu Y, Wang Z, Zheng J, Wang J, Chen Y, Huang C, Zhou H. RNA-seq revealed the anti-pyroptotic effect of suramin by suppressing NLRP3/caspase-1/GSDMD pathway in LPS-induced MH-S alveolar macrophages. Gene 2024; 893:147888. [PMID: 37839766 DOI: 10.1016/j.gene.2023.147888] [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: 06/15/2023] [Revised: 10/01/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), acting as one common sepsis-associated organ injury, induces uncontrolled and self-amplifies pulmonary inflammation. Given the lack of clinically effective approaches, the mortality rate of it still remains high. Suramin(SUR), as an antiparasitic drug initially, was found to ameliorate sepsis associated ALI in our previous work. However, the underlying mechanism of its protective effects has not been clarified. Pyroptosis, categorized as an inflammatory form of programmed cell death, could aggravate lung inflammatory responses via inducing alveolar macrophages (AM) pyroptosis. METHODS MH-S AM cell line was stimulated with or without lipopolysaccharide (LPS) or suramin, and the differential expression genes (DEGs) were excavated using RNA sequencing (RNA-seq). To identify the regulatory roles of these genes, pyroptosis-related genes (PRGs), GO/KEGG and GSEA analysis were conducted. We also performed WB, qRTPCR and ELISA to validate the RNA-seq results and further expound the protective effect of suramin. RESULTS 624 DEGs were identified between control (CON) and lipopolysaccharide (LPS) groups, and enrichment analysis of these genes revealed significantly enriched pathways that related to immune system and signal transduction. Meanwhile, 500 DEGs were identified in LPS/SUR+LPS group. In addition to the pathways mentioned above, IL-17 pathway and C-type lectin receptor signaling pathway were also enriched. All 6 pathways were connected with pyroptosis. Concurrently, the "DESeq2" R package was used to identify differentially expressed PRGs. Nod1, Nod2, interleukin (IL)-1b, IL-6, tumor necrosis factor (TNF), NLRP3 were upregulated under LPS stimulation. Then, in SUR+LPS group, Nod2, IL-6, IL-1b, NLRP3 were downregulated. The validation results of WB, qRT-PCR, and ELISA showed: the protein and mRNA expression levels of NLRP3, caspase-1, GSDMD and the concentrations of IL-1b, IL-18 were decreased when treated with suramin and LPS. CONCLUSION Suramin could inhibit NLRP3/caspase-1/GSDMD canonical pyroptosis pathway in LPS-induced MH-S alveolar macrophages.
Collapse
Affiliation(s)
- Yuhui Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Zhen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Jungang Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Jun Wang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Yijun Chen
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Changshun Huang
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Haidong Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, No. 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China.
| |
Collapse
|
67
|
Du X, Ren B, Li C, Li Q, Kan S, Wang X, Bai W, Wu C, Kassegne K, Yan H, Niu X, Yan M, Xu W, Wassmer SC, Wang J, Chen G, Wang Z. PRL2 regulates neutrophil extracellular trap formation which contributes to severe malaria and acute lung injury. Nat Commun 2024; 15:881. [PMID: 38286811 PMCID: PMC10825202 DOI: 10.1038/s41467-024-45210-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/17/2024] [Indexed: 01/31/2024] Open
Abstract
Excessive host immune responses contribute to severe malaria with high mortality. Here, we show that PRL2 in innate immune cells is highly related to experimental malaria disease progression, especially the development of murine severe malaria. In the absence of PRL2 in myeloid cells, Plasmodium berghei infection results in augmented lung injury, leading to significantly increased mortality. Intravital imaging revealed greater neutrophilic inflammation and NET formation in the lungs of PRL2 myeloid conditional knockout mice. Depletion of neutrophils prior to the onset of severe disease protected mice from NETs associated lung injury, and eliminated the difference between WT and PRL2 CKO mice. PRL2 regulates neutrophil activation and NET accumulation via the Rac-ROS pathway, thus contributing to NETs associated ALI. Hydroxychloroquine, an inhibitor of PRL2 degradation alleviates NETs associated tissue damage in vivo. Our findings suggest that PRL2 serves as an indicator of progression to severe malaria and ALI. In addition, our study indicated the importance of PRL2 in NET formation and tissue injury. It might open a promising path for adjunctive treatment of NET-associated disease.
Collapse
Affiliation(s)
- Xinyue Du
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, China; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Baiyang Ren
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, P.R. China
| | - Chang Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, P.R. China
| | - Qi Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Shuo Kan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Xin Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, P.R. China
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Chenyun Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Kokouvi Kassegne
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, China; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Huibo Yan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Xiaoyin Niu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Min Yan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500, P.R. China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University (The Third Military Medical University), Chongqing, 400038, P.R. China
| | - Samuel C Wassmer
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Jing Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China.
| | - Guangjie Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China.
| | - Zhaojun Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China.
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, China; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P.R. China.
| |
Collapse
|
68
|
Xuan N, Zhao J, Kang Z, Cui W, Tian BP. Neutrophil extracellular traps and their implications in airway inflammatory diseases. Front Med (Lausanne) 2024; 10:1331000. [PMID: 38283037 PMCID: PMC10811107 DOI: 10.3389/fmed.2023.1331000] [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: 10/31/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are essential for immune defense and have been increasingly recognized for their role in infection and inflammation. In the context of airway inflammatory diseases, there is growing evidence suggesting the involvement and significance of NETs. This review aims to provide an overview of the formation mechanisms and components of NETs and their impact on various airway inflammatory diseases, including acute lung injury/ARDS, asthma, chronic obstructive pulmonary disease (COPD) and cystic fibrosis. By understanding the role of NETs in airway inflammation, we can gain valuable insights into the underlying pathogenesis of these diseases and identify potential targets for future therapeutic strategies that either target NETs formation or modulate their harmful effects. Further research is warranted to elucidate the complex interactions between NETs and airway inflammation and to develop targeted therapies that can effectively mitigate their detrimental effects while preserving their beneficial functions in host defense.
Collapse
Affiliation(s)
- Nanxia Xuan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zhiying Kang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Cui
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bao-ping Tian
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
69
|
Fowler AA. Vitamin C: Rationale for Its Use in Sepsis-Induced Acute Respiratory Distress Syndrome (ARDS). Antioxidants (Basel) 2024; 13:95. [PMID: 38247519 PMCID: PMC10812524 DOI: 10.3390/antiox13010095] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening event that occurs in patients suffering from bacterial, fungal, or viral sepsis. Research performed over the last five decades showed that ARDS is a consequence of severe unrestrained systemic inflammation, which leads to injury of the lung's microvasculature and alveolar epithelium. ARDS leads to acute hypoxic/hypercapnic respiratory failure and death in a significant number of patients hospitalized in intensive care units worldwide. Basic and clinical research performed during the time since ARDS was first described has been unable to construct a pharmacological agent that will combat the inflammatory fire leading to ARDS. In-depth studies of the molecular pharmacology of vitamin C indicate that it can serve as a potent anti-inflammatory agent capable of attenuating the pathobiological events that lead to acute injury of the lungs and other body organs. This analysis of vitamin C's role in the treatment of ARDS includes a focused systematic review of the literature relevant to the molecular physiology of vitamin C and to the past performance of clinical trials using the agent.
Collapse
Affiliation(s)
- Alpha A Fowler
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
70
|
Zhang X, Su L, Pan P. Advances and Applications of Lung Organoids in the Research on Acute Respiratory Distress Syndrome (ARDS). J Clin Med 2024; 13:346. [PMID: 38256480 PMCID: PMC10816077 DOI: 10.3390/jcm13020346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a sudden onset of lung injury characterized by bilateral pulmonary edema, diffuse inflammation, hypoxemia, and a low P/F ratio. Epithelial injury and endothelial injury are notable in the development of ARDS, which is more severe under mechanical stress. This review explains the role of alveolar epithelial cells and endothelial cells under physiological and pathological conditions during the progression of ARDS. Mechanical injury not only causes ARDS but is also a side effect of ventilator-supporting treatment, which is difficult to model both in vitro and in vivo. The development of lung organoids has seen rapid progress in recent years, with numerous promising achievements made. Multiple types of cells and construction strategies are emerging in the lung organoid culture system. Additionally, the lung-on-a-chip system presents a new idea for simulating lung diseases. This review summarizes the basic features and critical problems in the research on ARDS, as well as the progress in lung organoids, particularly in the rapidly developing microfluidic system-based organoids. Overall, this review provides valuable insights into the three major factors that promote the progression of ARDS and how advances in lung organoid technology can be used to further understand ARDS.
Collapse
Affiliation(s)
- Xingwu Zhang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing 100091, China;
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Longxiang Su
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| | - Pan Pan
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing 100091, China;
| |
Collapse
|
71
|
Sapra L, Saini C, Das S, Mishra PK, Singh A, Mridha AR, Yadav PK, Srivastava RK. Lactobacillus rhamnosus (LR) ameliorates pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) via targeting neutrophils. Clin Immunol 2024; 258:109872. [PMID: 38113963 DOI: 10.1016/j.clim.2023.109872] [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: 08/11/2023] [Revised: 11/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) is a life-threatening respiratory failure associated with high mortality. Despite progress in our understanding of the pathological mechanism causing the crippling illness, there are currently no targeted pharmaceutical treatments available for it. Recent discoveries have emphasized the existence of a potential nexus between gut and lung health fueling novel approaches including probiotics for the treatment of ARDS. We thus investigated the prophylactic-potential of Lactobacillus rhamnosus-(LR) in lipopolysaccharide (LPS)-induced pulmonary and cecal ligation puncture (CLP) induced extrapulmonary ARDS mice. Our in-vivo findings revealed that pretreatment with LR significantly ameliorated vascular-permeability (edema) of the lungs via modulating the neutrophils along with significantly reducing the expression of inflammatory-cytokines in the BALF, lungs and serum in both pulmonary and extrapulmonary mice-models. Interestingly, our ex-vivo immunofluorescence and flow cytometric data suggested that mechanistically LR via short chain fatty acids (butyrate being the most potent and efficient in ameliorating the pathophysiology of both pulmonary and extra-pulmonary ARDS) targets the phagocytic and neutrophils extracellular traps (NETs) releasing potential of neutrophils. Moreover, our in-vivo data further corroborated our ex-vivo findings and suggested that butyrate exhibits enhanced potential in ameliorating the pathophysiology of ARDS via reducing the infiltration of neutrophils into the lungs. Altogether, our study establishes the prophylactic role of LR and its associated metabolites in the prevention and management of both pulmonary and extrapulmonary ARDS via targeting neutrophils.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sneha Das
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Anurag Singh
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Asit R Mridha
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pardeep K Yadav
- Central Animal Facility, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
72
|
Kosyreva AM, Miroshnichenko EA, Makarova OV. [The role of neutrophils in the mechanisms of sepsis]. Arkh Patol 2024; 86:82-91. [PMID: 39686902 DOI: 10.17116/patol20248606182] [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: 12/18/2024]
Abstract
The international consensus Sepsis-3 in 2016 defined sepsis as life-threatening organ dysfunction caused by a disruption of the host response to infection. One of the main mechanisms leading to the death of patients with sepsis is an imbalance of the immune response to pathogens. Activation of immune cells, in particular neutrophils, plays a key role in the mechanisms of sepsis. During sepsis, the number of immature neutrophils in the blood increases, PD-L1 neutrophils appear, which have an immunosuppressive function, and the number of Neu1 subtype of neutrophils, characterized by high expression of the NF-kB inhibitor, the chemokine CXCL8, olfactomedine, which regulates the production of cytokines, and myeloperoxidase, increases. When the inflammatory response generalizes, the phagocytic and bactericidal functions of neutrophils are disrupted. An important role in the mechanisms of sepsis is played by the formation of neutrophil extracellular traps, extracellular DNA, and the nuclear non-histone protein HMGB1, which are DAMPs and have proinflammatory activity. The development of approaches to predicting the development and course of sepsis in injuries and infectious and inflammatory diseases based on assessing the functional activity of neutrophils and their phenotype is a promising area of research, in particular approaches to regulating the functioning of neutrophils using miRNA.
Collapse
Affiliation(s)
- A M Kosyreva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba, Moscow, Russia
| | - E A Miroshnichenko
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba, Moscow, Russia
| | - O V Makarova
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| |
Collapse
|
73
|
Nagaraju S, Ramalingam S, Mani S. Pulmonary Manifestations of COVID-19. TEXTBOOK OF SARS-COV-2 AND COVID-19 2024:100-136. [DOI: 10.1016/b978-0-323-87539-4.00005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
74
|
Sun B, Lei M, Zhang J, Kang H, Liu H, Zhou F. Acute lung injury caused by sepsis: how does it happen? Front Med (Lausanne) 2023; 10:1289194. [PMID: 38076268 PMCID: PMC10702758 DOI: 10.3389/fmed.2023.1289194] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2024] Open
Abstract
Sepsis is a systemic inflammatory disease caused by severe infections that involves multiple systemic organs, among which the lung is the most susceptible, leaving patients highly vulnerable to acute lung injury (ALI). Refractory hypoxemia and respiratory distress are classic clinical symptoms of ALI caused by sepsis, which has a mortality rate of 40%. Despite the extensive research on the mechanisms of ALI caused by sepsis, the exact pathological process is not fully understood. This article reviews the research advances in the pathogenesis of ALI caused by sepsis by focusing on the treatment regimens adopted in clinical practice for the corresponding molecular mechanisms. This review can not only contribute to theories on the pathogenesis of ALI caused by sepsis, but also recommend new treatment strategies for related injuries.
Collapse
Affiliation(s)
- Baisheng Sun
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Mingxing Lei
- Department of Orthopedic Surgery, Hainan Hospital of Chinese PLA General Hospital, Beijing, China
- Department of Orthopedic Surgery, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jiaqi Zhang
- Medical School of Chinese PLA, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hui Liu
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Feihu Zhou
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Medical Engineering Laboratory of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
75
|
Lim EY, Lee SY, Shin HS, Kim GD. Reactive Oxygen Species and Strategies for Antioxidant Intervention in Acute Respiratory Distress Syndrome. Antioxidants (Basel) 2023; 12:2016. [PMID: 38001869 PMCID: PMC10669909 DOI: 10.3390/antiox12112016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening pulmonary condition characterized by the sudden onset of respiratory failure, pulmonary edema, dysfunction of endothelial and epithelial barriers, and the activation of inflammatory cascades. Despite the increasing number of deaths attributed to ARDS, a comprehensive therapeutic approach for managing patients with ARDS remains elusive. To elucidate the pathological mechanisms underlying ARDS, numerous studies have employed various preclinical models, often utilizing lipopolysaccharide as the ARDS inducer. Accumulating evidence emphasizes the pivotal role of reactive oxygen species (ROS) in the pathophysiology of ARDS. Both preclinical and clinical investigations have asserted the potential of antioxidants in ameliorating ARDS. This review focuses on various sources of ROS, including NADPH oxidase, uncoupled endothelial nitric oxide synthase, cytochrome P450, and xanthine oxidase, and provides a comprehensive overview of their roles in ARDS. Additionally, we discuss the potential of using antioxidants as a strategy for treating ARDS.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| |
Collapse
|
76
|
Xu H, Sheng S, Luo W, Xu X, Zhang Z. Acute respiratory distress syndrome heterogeneity and the septic ARDS subgroup. Front Immunol 2023; 14:1277161. [PMID: 38035100 PMCID: PMC10682474 DOI: 10.3389/fimmu.2023.1277161] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute diffuse inflammatory lung injury characterized by the damage of alveolar epithelial cells and pulmonary capillary endothelial cells. It is mainly manifested by non-cardiogenic pulmonary edema, resulting from intrapulmonary and extrapulmonary risk factors. ARDS is often accompanied by immune system disturbance, both locally in the lungs and systemically. As a common heterogeneous disease in critical care medicine, researchers are often faced with the failure of clinical trials. Latent class analysis had been used to compensate for poor outcomes and found that targeted treatment after subgrouping contribute to ARDS therapy. The subphenotype of ARDS caused by sepsis has garnered attention due to its refractory nature and detrimental consequences. Sepsis stands as the most predominant extrapulmonary cause of ARDS, accounting for approximately 32% of ARDS cases. Studies indicate that sepsis-induced ARDS tends to be more severe than ARDS caused by other factors, leading to poorer prognosis and higher mortality rate. This comprehensive review delves into the immunological mechanisms of sepsis-ARDS, the heterogeneity of ARDS and existing research on targeted treatments, aiming to providing mechanism understanding and exploring ideas for accurate treatment of ARDS or sepsis-ARDS.
Collapse
Affiliation(s)
- Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiying Sheng
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weiwei Luo
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China
- Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| |
Collapse
|
77
|
Wu Y, Wang H, Song A, Wang X, Ma Q, Yao C, Xu J, Dai H, Wang C, Lu T, Xu F. PD-L1-Expressing Extracellular Vesicles for the Treatment of Pneumonia. ACS Biomater Sci Eng 2023; 9:6464-6471. [PMID: 37844209 DOI: 10.1021/acsbiomaterials.3c01173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a severe lung condition with a high mortality rate and a lack of effective drug therapy. In this work, we developed mesenchymal stem cell (MSC)-derived extracellular vesicles with high PD-L1 expression (MSC-EVs-PD-L1) for treating lipopolysaccharide (LPS)-induced pneumonia by intratracheal administration. We found an upregulation of PD-1 expression in the inflammatory region of murine lungs; hence, MSC-EVs-PD-L1 exerted immunosuppressive effects via the PD-1/PD-L1 signaling pathway. Furthermore, we treated LPS-induced pneumonia mice by intratracheal administration, which enabled heavy drug accumulation in the lungs of mice and better therapeutic efficacy compared to systemic administration. Our results suggest that MSC-EVs-PD-L1 has the potential to provide a universal platform technology for the immunotherapy of pneumonia.
Collapse
Affiliation(s)
- Yi Wu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Department of Biochemistry and Molecular Biology, Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Heng Wang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Anning Song
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xiaoyu Wang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Chenlu Yao
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Ting Lu
- Department of Biochemistry and Molecular Biology, Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
78
|
Schmidt C, Weißmüller S, Heinz CC. Multifaceted Tissue-Protective Functions of Polyvalent Immunoglobulin Preparations in Severe Infections-Interactions with Neutrophils, Complement, and Coagulation Pathways. Biomedicines 2023; 11:3022. [PMID: 38002022 PMCID: PMC10669904 DOI: 10.3390/biomedicines11113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Severe infections induce immune defense mechanisms and initial tissue damage, which produce an inflammatory neutrophil response. Upon dysregulation of these responses, inflammation, further tissue damage, and systemic spread of the pathogen may occur. Subsequent vascular inflammation and activation of coagulation processes may cause microvascular obstruction at sites distal to the primary site of infection. Low immunoglobulin (Ig) M and IgG levels have been detected in patients with severe infections like sCAP and sepsis, associated with increased severity and mortality. Based on Ig's modes of action, supplementation with polyvalent intravenous Ig preparations (standard IVIg or IgM/IgA-enriched Ig preparations) has long been discussed as a treatment option for severe infections. A prerequisite seems to be the timely administration of Ig preparations before excessive tissue damage has occurred and coagulopathy has developed. This review focuses on nonclinical and clinical studies that evaluated tissue-protective activities resulting from interactions of Igs with neutrophils, complement, and the coagulation system. The data indicate that coagulopathy, organ failure, and even death of patients can possibly be prevented by the timely combined interactions of (natural) IgM, IgA, and IgG with neutrophils and complement.
Collapse
Affiliation(s)
- Carolin Schmidt
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| | | | - Corina C Heinz
- Department of Corporate Clinical Research and Development, Biotest AG, 63303 Dreieich, Germany
| |
Collapse
|
79
|
Shen L, Cai N, Wan S, Chen S. Development and validation of a predictive model for early diagnosis of neonatal acute respiratory distress syndrome based on the Montreux definition. Front Pediatr 2023; 11:1276915. [PMID: 38027256 PMCID: PMC10652555 DOI: 10.3389/fped.2023.1276915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Based on the Montreux definition, we aim to develop and validate a predictive model for the early diagnosis of neonatal acute respiratory distress syndrome (ARDS). Methods A retrospective analysis of clinical data on 198 neonates with respiratory distress from January 2018 to January 2022 was conducted. Neonates meeting Montreux definition were classified as ARDS group (n = 79), while the rest were non-ARDS group (n = 119). Univariate analysis identified indicators for neonatal ARDS, followed by logistic regression to construct a predictive model for early diagnosis. The ability of predictors and models to predict neonatal ARDS was evaluated using area under the curve (AUC), and model performance was estimated through bootstrap resampling. Results Maternal prenatal fever, abnormal fetal heart beat, meconium-stained amniotic fluid (MSAF), white blood cell (WBC), absolute neutrophil count (ANC), neutrophil percentage (NE%), platelet count (PLT), C-reactive protein (CRP), procalcitonin (PCT), creatine kinase (CK), activated partial thromboplastin time (APTT), serum calcium (Ca) and sodium (Na)exhibited significant differences between the ARDS group and the non-ARDS group (P < 0.05). MSAF (OR=5.037; 95% CI: 1.523-16.657; P < 0.05), ANC (OR = 1.324; 95% CI: 1.172-1.495; P < 0.05), PLT (OR = 0.979; 95% CI: 0.971-0.986; P < 0.05), Ca (OR = 0.020; 95% CI: 0.004-0.088; P < 0.05) emerged as independent risk factors for the development of ARDS. The respective AUC values for MSAF, ANC, PLT, Ca, and the combined prediction models were 0.606, 0.691, 0.808, 0.761 and 0.931. Internal validation showed that the C-index for the model was 0.931. Conclusions Early application of the model combining MSAF, ANC, PLT and Ca may have a good predictive effect on the early diagnosis of neonatal ARDS.
Collapse
Affiliation(s)
| | | | | | - Sheng Chen
- Department of Pediatrics, The First Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
80
|
Lingel MP, Haus M, Paschke L, Foltan M, Lubnow M, Gruber M, Krenkel L, Lehle K. Clinical relevance of cell-free DNA during venovenous extracorporeal membrane oxygenation. Artif Organs 2023; 47:1720-1731. [PMID: 37525949 DOI: 10.1111/aor.14616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Thrombosis remains a critical complication during venovenous extracorporeal membrane oxygenation (VV ECMO). The involvement of neutrophil extracellular traps (NETs) in thrombogenesis has to be discussed. The aim was to verify NETs in the form of cell-free DNA (cfDNA) in the plasma of patients during ECMO. METHODS A fluorescent DNA-binding dye (QuantifFluor®, Promega) was used to detect cell-free DNA in plasma samples. cfDNA concentrations from volunteers (n = 21) and patients (n = 9) were compared and correlated with clinical/technical data before/during support, ECMO end and time of a system exchange. RESULTS Before ECMO, patients with a median (IQR) age of 59 (51/63) years, SOFA score of 11 (10/15), and ECMO run time of 9.0 (7.0/19.5) days presented significantly higher levels of cfDNA compared to volunteers (6.4 (5.8/7.9) ng/μL vs. 5.9 (5.4/6.3) ng/μL; p = 0.044). Within 2 days after ECMO start, cfDNA, inflammatory, and hemolysis parameters remained unchanged, while platelets decreased (p = 0.005). After ECMO removal at the end of therapy, cfDNA, inflammation, and coagulation data (except antithrombin III) remained unchanged. The renewal of a system resulted in known alterations in fibrinogen, d-dimers, and platelets, while cfDNA remained unchanged. CONCLUSION Detection of cfDNA in plasma of ECMO patients was not an indicator of acute and circuit-induced thrombogenesis.
Collapse
Affiliation(s)
- Maximilian P Lingel
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Moritz Haus
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Paschke
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Maik Foltan
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Lubnow
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Lars Krenkel
- Regensburg Center of Biomedical Engineering, Ostbayerische Technische Hochschule, Regensburg, Germany
| | - Karla Lehle
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
81
|
Banerji R, Grifno GN, Shi L, Smolen D, LeBourdais R, Muhvich J, Eberman C, Hiller BE, Lee J, Regan K, Zheng S, Zhang S, Jiang J, Raslan AA, Breda JC, Pihl R, Traber K, Mazzilli S, Ligresti G, Mizgerd JP, Suki B, Nia HT. Crystal ribcage: a platform for probing real-time lung function at cellular resolution. Nat Methods 2023; 20:1790-1801. [PMID: 37710017 PMCID: PMC10860663 DOI: 10.1038/s41592-023-02004-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 08/10/2023] [Indexed: 09/16/2023]
Abstract
Understanding the dynamic pathogenesis and treatment response in pulmonary diseases requires probing the lung at cellular resolution in real time. Despite advances in intravital imaging, optical imaging of the lung during active respiration and circulation has remained challenging. Here, we introduce the crystal ribcage: a transparent ribcage that allows multiscale optical imaging of the functioning lung from whole-organ to single-cell level. It enables the modulation of lung biophysics and immunity through intravascular, intrapulmonary, intraparenchymal and optogenetic interventions, and it preserves the three-dimensional architecture, air-liquid interface, cellular diversity and respiratory-circulatory functions of the lung. Utilizing these capabilities on murine models of pulmonary pathologies we probed remodeling of respiratory-circulatory functions at the single-alveolus and capillary levels during disease progression. The crystal ribcage and its broad applications presented here will facilitate further studies of nearly any pulmonary disease as well as lead to the identification of new targets for treatment strategies.
Collapse
Affiliation(s)
- Rohin Banerji
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Gabrielle N Grifno
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Linzheng Shi
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dylan Smolen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rob LeBourdais
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Johnathan Muhvich
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cate Eberman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Bradley E Hiller
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kathryn Regan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Siyi Zheng
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - John Jiang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ahmed A Raslan
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Julia C Breda
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Riley Pihl
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Katrina Traber
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sarah Mazzilli
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Giovanni Ligresti
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
82
|
Guilherme Neto JL, Rodrigues Venturini LG, Schneider AH, Taira TM, Duffles Rodrigues LF, Veras FP, Oliveira SR, da Silva TA, Cunha FQ, Fukada SY. Neutrophil Extracellular Traps Aggravate Apical Periodontitis by Stimulating Osteoclast Formation. J Endod 2023; 49:1514-1521. [PMID: 37619708 DOI: 10.1016/j.joen.2023.07.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
INTRODUCTION Neutrophil extracellular traps (NETs) have been described as structures composed of DNA and proteins, such as elastase and myeloperoxidase, that are able to kill bacteria extracellularly. The aim of the present study was to evaluate the role of NETs in bone resorption observed in pulp infection-induced apical periodontitis in mice. METHODS Apical periodontitis was experimentally induced by exposing the dental pulp of the mandibular first molar of mice to the oral microenvironment. The expression of NETs was evaluated by immunofluorescence in mice and biopsies of apical periodontitis. Mice were treated with vehicle or DNase I to degrade NETs, and the samples were collected after 7 days. The size of the apical lesion and the osteoclast number were determined in hematoxylin-eosin- and tartrate-resistant acid phosphatase-stained sections, respectively. Osteoclast differentiation and function markers were evaluated by quantitative polymerase chain reaction. The level of NETs in the serum was determined by the myeloperoxidase-DNA PicoGreen assay. RESULTS We first confirmed the presence of neutrophils and NETs at the site of the lesion in mice and in biopsies of patients with apical periodontitis. The treatment of mice with DNase I reduced the level of NETs in the serum and led to a reduction in apical lesion size and alveolar bone resorption. This effect was associated with a reduction of local inflammatory infiltrate and a reduced number of osteoclasts. We found that the increased expression of Acp5, Ctsk, and Rankl genes associated with osteoclast formation and function were abrogated by the absence of NETs. CONCLUSIONS Our data highlight NETs as an important player in the pathogenesis of apical periodontitis with regard to the local inflammation and consequent bone resorption after pulp infection.
Collapse
Affiliation(s)
- João Luiz Guilherme Neto
- Laboratory of Bone Biology, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Gabriel Rodrigues Venturini
- Laboratory of Bone Biology, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ayda Henriques Schneider
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thaise Mayumi Taira
- Laboratory of Bone Biology, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Letícia Fernanda Duffles Rodrigues
- Laboratory of Bone Biology, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Flávio Protásio Veras
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sicília Rezende Oliveira
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Dentistry School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tarcília Aparecida da Silva
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Dentistry School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sandra Yasuyo Fukada
- Laboratory of Bone Biology, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
83
|
Yadav R, Momin A, Godugu C. DNase based therapeutic approaches for the treatment of NETosis related inflammatory diseases. Int Immunopharmacol 2023; 124:110846. [PMID: 37634446 DOI: 10.1016/j.intimp.2023.110846] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/19/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
Neutrophils are the primary host innate immune cells defending against pathogens. One proposed mechanism by which neutrophils limit pathogen transmission is NETosis, which includes releasing the nuclear content into the cytosol by forming pores in the plasma membrane. The extrusion of cellular deoxyribonucleic acid (DNA) results in neutrophil extracellular traps (NETs) composed of nuclear DNA associated with histones and granule proteins. NETosis is driven by the enzyme PAD-4 (Peptidylarginine deiminase-4), which converts arginine into citrulline, leading to decondensation of chromatin, separation of DNA, and eventual extrusion. DNase is responsible for the breakdown of NETs. On the one hand, the release of DNase may interfere with the antibacterial effects of NETs; further, DNase may protect tissues from self-destruction caused by the increased release of NET under septic conditions. NETs in physiological quantities are expected to have a role in anti-infectious innate immune responses. In contrast, abnormally high concentrations of NETs in the body that are not adequately cleared by DNases can damage tissues and cause inflammation. Through several novel approaches, it is now possible to avoid the adverse effects caused by the continued release of NETs into the extracellular environment. In this review we have highlighted the basic mechanisms of NETosis, its significance in the pathogenesis of various inflammatory disorders, and the role of DNase enzyme with a focus on the possible function of nanotechnology in its management.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Alfiya Momin
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
84
|
Islam MM, Takeyama N. Role of Neutrophil Extracellular Traps in Health and Disease Pathophysiology: Recent Insights and Advances. Int J Mol Sci 2023; 24:15805. [PMID: 37958788 PMCID: PMC10649138 DOI: 10.3390/ijms242115805] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Neutrophils are the principal trouper of the innate immune system. Activated neutrophils undergo a noble cell death termed NETosis and release a mesh-like structure called neutrophil extracellular traps (NETs) as a part of their defensive strategy against microbial pathogen attack. This web-like architecture includes a DNA backbone embedded with antimicrobial proteins like myeloperoxidase (MPO), neutrophil elastase (NE), histones and deploys in the entrapment and clearance of encountered pathogens. Thus NETs play an inevitable beneficial role in the host's protection. However, recent accumulated evidence shows that dysregulated and enhanced NET formation has various pathological aspects including the promotion of sepsis, pulmonary, cardiovascular, hepatic, nephrological, thrombotic, autoimmune, pregnancy, and cancer diseases, and the list is increasing gradually. In this review, we summarize the NET-mediated pathophysiology of different diseases and focus on some updated potential therapeutic approaches against NETs.
Collapse
Affiliation(s)
- Md Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Aichi 480-1195, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram 4202, Bangladesh
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Aichi 480-1195, Japan
| |
Collapse
|
85
|
Tu F, Pan L, Wu W, Cai Y, Li J, Wang X, Lai X, Chen Z, Ye L, Wang S. Recombinant GM-CSF enhances the bactericidal ability of PMNs by increasing intracellular IL-1β and improves the prognosis of secondary Pseudomonas aeruginosa pneumonia in sepsis. J Leukoc Biol 2023; 114:443-458. [PMID: 37490847 DOI: 10.1093/jleuko/qiad088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023] Open
Abstract
This study tested the hypothesis that recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances polymorphonuclear neutrophils (PMNs) via interleukin (IL)-1β to improve the prognosis of secondary infection in sepsis. The latter stage of sepsis is prone to induce immunosuppression, resulting in secondary fatal infections. Recombinant GM-CSF has become a way for sepsis-induced immunosuppression due to its immunomodulatory effect. However, the functional impact of GM-CSF on PMNs in sepsis remains obscure. This study aimed to study the role of recombinant GM-CSF on the bactericidal ability of PMNs in septic mice, assessing its effect on the prognosis of secondary pneumonia, and explore the mechanism of recombinant GM-CSF by intervening PMNs in patients with sepsis. The C57BL/6J sepsis mouse model was induced by cecal ligation and puncture. Recombinant murine GM-CSF (rmGM-CSF) was used in vivo when mice developed immunosuppression, which was characterized by abnormal bactericidal function of PMNs in peripheral blood. rmGM-CSF improved the prognosis of secondary pneumonia and reversed the function of PMNs. PMNs isolated by Percoll from septic patients were treated by recombinant human GM-CSF (rhGM-CSF) in vitro. The expression of CD11b, reactive oxygen species, phagocytosis, and neutrophil extracellular trap release in PMNs were enhanced by rhGM-CSF treatments. Whole-transcriptomic sequencing of mouse PMNs indicated that recombinant GM-CSF increased the expression of Il1b gene in PMNs. Blocking and inhibiting IL-1β release effectively counteracted the enhancing effect of GM-CSF on the bactericidal function of PMNs. rmGM-CSF enhances the bactericidal function of PMNs in vivo and improves the prognosis of secondary pneumonia in septic mice, and recombinant GM-CSF increases IL-1β precursor reserves, which, if stimulated, can rapidly enhance the bactericidal capacity of PMNs.
Collapse
Affiliation(s)
- Fuquan Tu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Department of Emergency Intensive Care Unit, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Lili Pan
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Wenwei Wu
- Department of Emergency Intensive Care Unit, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Yuanhua Cai
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Jinggang Li
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Xuechun Wang
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Xiaolin Lai
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Zhixiang Chen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Luya Ye
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| | - Shaoyuan Wang
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Department of Emergency Intensive Care Unit, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, Fujian, China
- Union Clinical Medical Colleges, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, Fujian, China
| |
Collapse
|
86
|
Yang X, Liu X, Nie Y, Zhan F, Zhu B. Oxidative stress and ROS-mediated cellular events in RSV infection: potential protective roles of antioxidants. Virol J 2023; 20:224. [PMID: 37798799 PMCID: PMC10557227 DOI: 10.1186/s12985-023-02194-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023] Open
Abstract
Respiratory syncytial virus (RSV), a member of the Pneumoviridae family, can cause severe acute lower respiratory tract infection in infants, young children, immunocompromised individuals and elderly people. RSV is associated with an augmented innate immune response, enhanced secretion of inflammatory cytokines, and necrosis of infected cells. Oxidative stress, which is mainly characterized as an imbalance in the production of reactive oxygen species (ROS) and antioxidant responses, interacts with all the pathophysiologic processes above and is receiving increasing attention in RSV infection. A gradual accumulation of evidence indicates that ROS overproduction plays an important role in the pathogenesis of severe RSV infection and serves as a major factor in pulmonary inflammation and tissue damage. Thus, antioxidants seem to be an effective treatment for severe RSV infection. This article mainly reviews the information on oxidative stress and ROS-mediated cellular events during RSV infection for the first time.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Xue Liu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Yujun Nie
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Fei Zhan
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Bin Zhu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China.
| |
Collapse
|
87
|
Peng W, Qi H, Zhu W, Tong L, Rouzi A, Wu Y, Han L, He L, Yan Y, Pan T, Liu J, Wang Q, Jia Z, Song Y, Zhu Q, Zhou J. Lianhua Qingke ameliorates lipopolysaccharide-induced lung injury by inhibiting neutrophil extracellular traps formation and pyroptosis. Pulm Circ 2023; 13:e12295. [PMID: 37808899 PMCID: PMC10557103 DOI: 10.1002/pul2.12295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023] Open
Abstract
LHQK is a patented Traditional Chinese Medicine (TCM) which is clinically used for acute tracheobronchitis, cough, and other respiratory diseases. Recent studies have proved that LHQK exhibits excellent clinical efficacy in the treatment of acute lung injury (ALI). However, the corresponding mechanisms remain largely unexplored. In this study, we investigated the effects and the underlying mechanisms of LHQK on lipopolysaccharide (LPS)-induced ALI in mice. The pathological examination, inflammatory cytokines assessments, and mucus secretion evaluation indicated that administration of LHQK ameliorated LPS-induced lung injury, and suppressed the secretion of Muc5AC and pro-inflammatory cytokines (IL-6, TNF-α, and IL-1β) in plasma and BALF. Furthermore, the results of cell-free DNA level showed that LHQK significantly inhibited LPS-induced NETs formation. Western blot revealed that LHQK effectively inhibited LPS-triggered pyroptosis in the lung. In addition, RNA-Seq data analysis, relatively bioinformatic analysis, and network pharmacology analysis revealed that LHQK and relative components may play multiple protective functions in LPS-induced ALI/acute respiratory distress syndrome (ARDS) by regulating multiple targets directly or indirectly related to NETs and pyroptosis. In conclusion, LHQK can effectively attenuate lung injury and reduce lung inflammation by inhibiting LPS-induced NETs formation and pyroptosis, which may be regulated directly or indirectly by active compounds of LHQK.
Collapse
Affiliation(s)
- Wenjun Peng
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western MedicineHebeiShijiazhuangChina
| | - Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Lin Tong
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ainiwaer Rouzi
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Yuanyuan Wu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Linxiao Han
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ludan He
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Yu Yan
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Ting Pan
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Jie Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
| | - Zhenhua Jia
- Hebei Academy of Integrated Traditional Chinese and Western MedicineHebeiShijiazhuangChina
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan UniversityFudan UniversityShanghaiChina
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan UniversityFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionFudan UniversityShanghaiChina
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan UniversityFudan UniversityShanghaiChina
| |
Collapse
|
88
|
Linnemann C, Sahin F, Chen Y, Falldorf K, Ronniger M, Histing T, Nussler AK, Ehnert S. NET Formation Was Reduced via Exposure to Extremely Low-Frequency Pulsed Electromagnetic Fields. Int J Mol Sci 2023; 24:14629. [PMID: 37834077 PMCID: PMC10572227 DOI: 10.3390/ijms241914629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Fracture-healing is a highly complex and timely orchestrated process. Non-healing fractures are still a major clinical problem and treatment remains difficult. A 16 Hz extremely low-frequency pulsed electromagnetic field (ELF-PEMF) was identified as non-invasive adjunct therapy supporting bone-healing by inducing reactive oxygen species (ROS) and Ca2+-influx. However, ROS and Ca2+-influx may stimulate neutrophils, the first cells arriving at the wounded site, to excessively form neutrophil extracellular traps (NETs), which negatively affects the healing process. Thus, this study aimed to evaluate the effect of this 16 Hz ELF-PEMF on NET formation. Neutrophils were isolated from healthy volunteers and exposed to different NET-stimuli and the 16 Hz ELF-PEMF. NETs were quantified using Sytox Green Assay and immunofluorescence, Ca2+-influx and ROS with fluorescence probes. In contrast to mesenchymal cells, ELF-PEMF exposure did not induce ROS and Ca2+-influx in neutrophils. ELF-PEMF exposure did not result in basal or enhanced PMA-induced NET formation but did reduce the amount of DNA released. Similarly, NET formation induced by LPS and H2O2 was reduced through exposure to ELF-PEMF. As ELF-PEMF exposure did not induce NET release or negatively affect neutrophils, the ELF-PEMF exposure can be started immediately after fracture treatment.
Collapse
Affiliation(s)
- Caren Linnemann
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Filiz Sahin
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Yangmengfan Chen
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Karsten Falldorf
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Michael Ronniger
- Sachtleben GmbH, Haus Spectrum am UKE, Martinistraße 64, 20251 Hamburg, Germany
| | - Tina Histing
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, Schnarrenbergstraße 95, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| |
Collapse
|
89
|
Fu D, Jiang J, Fu S, Xie D, Gao C, Feng Y, Liu S, Ye Y, Liu L, Tu Y, Peng F. Real-Time Micromotor Probe for Immune Neutrophil Activation State. Adv Healthc Mater 2023; 12:e2300737. [PMID: 37199571 DOI: 10.1002/adhm.202300737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Indexed: 05/19/2023]
Abstract
Neutrophil activation is a hallmark of the immune response. Approaches to identify neutrophil activation in real time are necessary but are still lacking. In this study, magnetic Spirulina micromotors are used as label-free probes that exhibit differences in motility under different neutrophil activation states. This is correlated with different secretions into the extracellular environment by activated/non-activated cells and local environmental viscoelasticity. The micromotor platform can bypass non-activated immune cells while being stopped by activated cells. Thus, the micromotors can serve as label-free biomechanical probes of the immune cell state. They can detect the activation state of target immune cells in real time and with single-cell precision, which provides new ideas for the diagnosis and treatment of diseases while deepening understanding of the biomechanics of activated immune cells.
Collapse
Affiliation(s)
- Dongmei Fu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jiamiao Jiang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shaoming Fu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Dazhi Xie
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Chao Gao
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Ye Feng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Suyi Liu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yicheng Ye
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yingfeng Tu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
90
|
Sikora JP, Karawani J, Sobczak J. Neutrophils and the Systemic Inflammatory Response Syndrome (SIRS). Int J Mol Sci 2023; 24:13469. [PMID: 37686271 PMCID: PMC10488036 DOI: 10.3390/ijms241713469] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
We are not entirely able to understand, assess, and modulate the functioning of the immune system in clinical situations that lead to a systemic inflammatory response. In the search for diagnostic and treatment strategies (which are still far from perfect), it became very important to study the pathogenesis and participation of endogenous inflammation mediators. This study attempts to more precisely establish the role of neutrophils in individual phenomena occurring during an inflammatory and anti-inflammatory reaction, taking into account their cidal, immunoregulatory, and reparative abilities. Pro- and anticoagulatory properties of endothelium in systemic inflammatory response syndrome (SIRS) are emphasised, along with the resulting clinical implications (the application of immunotherapy using mesenchymal stem/stromal cells (MSCs) or IL-6 antagonists in sepsis and COVID-19 treatment, among others). Special attention is paid to reactive oxygen species (ROS), produced by neutrophils activated during "respiratory burst" in the course of SIRS; the protective and pathogenic role of these endogenous mediators is highlighted. Moreover, clinically useful biomarkers of SIRS (neutrophil extracellular traps, cell-free DNA, DAMP, TREMs, NGAL, miRNA, selected cytokines, ROS, and recognised markers of endothelial damage from the group of adhesins by means of immunohistochemical techniques) related to the neutrophils are presented, and their role in the diagnosing and forecasting of sepsis, burn disease, and COVID-19 is emphasised. Finally, examples of immunomodulation of sepsis and antioxidative thermal injury therapy are presented.
Collapse
Affiliation(s)
- Janusz P. Sikora
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
| | - Jakub Karawani
- Faculty of Medicine, Lazarski University, ul. Świeradowska 43, 02-662 Warsaw, Poland;
| | - Jarosław Sobczak
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
- Department of Management and Logistics in Healthcare, Medical University of Łódź, ul. Lindleya 6, 90-131 Łódź, Poland
| |
Collapse
|
91
|
Huber A, Jose S, Kassam A, Weghorn KN, Powers-Fletcher M, Sharma D, Mukherjee A, Mathew A, Kulkarni N, Chandramouli S, Alder MN, Madan R. Olfactomedin-4 + neutrophils exacerbate intestinal epithelial damage and worsen host survival after Clostridioides difficile infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.553751. [PMID: 37662327 PMCID: PMC10473617 DOI: 10.1101/2023.08.21.553751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Neutrophils are key first responders to Clostridioides difficile infection (CDI). Excessive tissue and blood neutrophils are associated with worse histopathology and adverse outcomes, however their functional role during CDI remains poorly defined. Utilizing intestinal epithelial cell (IEC)-neutrophil co-cultures and a pre-clinical animal model of CDI, we show that neutrophils exacerbate C. difficile -induced IEC injury. We utilized cutting-edge single-cell transcriptomics to illuminate neutrophil subtypes and biological pathways that could exacerbate CDI-associated IEC damage. As such, we have established the first transcriptomics atlas of bone marrow (BM), blood, and colonic neutrophils after CDI. We found that CDI altered the developmental trajectory of BM and blood neutrophils towards populations that exhibit gene signatures associated with pro-inflammatory responses and neutrophil-mediated tissue damage. Similarly, the transcriptomic signature of colonic neutrophils was consistent with hyper-inflammatory and highly differentiated cells that had amplified expression of cytokine-mediated signaling and degranulation priming genes. One of the top 10 variable features in colonic neutrophils was the gene for neutrophil glycoprotein, Olfactomedin 4 (OLFM4). CDI enhanced OLFM4 mRNA and protein expression in neutrophils, and OLFM4 + cells aggregated to areas of severe IEC damage. Compared to uninfected controls, both humans and mice with CDI had higher concentrations of circulating OLFM4; and in mice, OLFM4 deficiency resulted in faster recovery and better survival after infection. Collectively, these studies provide novel insights into neutrophil-mediated pathology after CDI and highlight the pathogenic role of OLFM4 + neutrophils in regulating CDI-induced IEC damage. One Sentence Summary Utilizing single-cell transcriptomics, IEC-epithelial co-cultures, and pre-clinical models of CDI, we have identified a subset of neutrophils that are marked by OLFM4 expression as pathogenic determinants of IEC barrier damage after CDI.
Collapse
|
92
|
Wu J, Lan Y, Wu J, Zhu K. Sepsis-Induced Acute Lung Injury Is Alleviated by Small Molecules from Dietary Plants via Pyroptosis Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:12153-12166. [PMID: 37537751 DOI: 10.1021/acs.jafc.2c08926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Sepsis-induced acute respiratory distress syndrome (ARDS) has high morbidity and mortality, and it has three major pathogeneses, namely alveolar-capillary barrier destruction, elevated gut permeability, and reduced neutrophil extracellular traps (NETS), all of which are pyroptosis-involved. Due to limitations of current agents like adverse reaction superposition, inevitable drug resistance, and relatively heavier financial burden, naturally extracted small-molecule compounds have a broad market even though chemically modified drugs have straightforward efficacy. Despite increased understanding of the molecular biology and mechanism underlying sepsis-induced ARDS, there are no specific reviews concerning how small molecules from dietary plants alleviate sepsis-induced acute lung injury (ALI) via regulating pyroptotic cell death. Herein, we traced and reviewed the molecular underpinnings of sepsis-induced ALI with a focus on small-molecule compounds from dietary plants, the top three categories of which are respectively flavonoids and flavone, terpenoids, and polyphenol and phenolic acids, and how they rescued septic ALI by restraining pyroptosis.
Collapse
Affiliation(s)
- Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuejia Lan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Jinghan Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Keli Zhu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
93
|
Pi P, Zeng Z, Zeng L, Han B, Bai X, Xu S. Molecular mechanisms of COVID-19-induced pulmonary fibrosis and epithelial-mesenchymal transition. Front Pharmacol 2023; 14:1218059. [PMID: 37601070 PMCID: PMC10436482 DOI: 10.3389/fphar.2023.1218059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
As the outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first broke out in Hubei Province, China, at the end of 2019. It has brought great challenges and harms to global public health. SARS-CoV-2 mainly affects the lungs and is mainly manifested as pulmonary disease. However, one of the biggest crises arises from the emergence of COVID-19-induced fibrosis. At present, there are still many questions about how COVID-19 induced pulmonary fibrosis (PF) occurs and how to treat and regulate its long-term effects. In addition, as an important process of fibrosis, the effect of COVID-19 on epithelial-mesenchymal transition (EMT) may be an important factor driving PF. This review summarizes the main pathogenesis and treatment mechanisms of COVID-19 related to PF. Starting with the basic mechanisms of PF, such as EMT, transforming growth factor-β (TGF-β), fibroblasts and myofibroblasts, inflammation, macrophages, innate lymphoid cells, matrix metalloproteinases and tissue inhibitors of metalloproteinases, hedgehog pathway as well as Notch signaling. Further, we highlight the importance of COVID-19-induced EMT in the process of PF and provide an overview of the related molecular mechanisms, which will facilitate future research to propose new clinical therapeutic solutions for the treatment of COVID-19-induced PF.
Collapse
Affiliation(s)
- Peng Pi
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Zhipeng Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Liqing Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Bing Han
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Xizhe Bai
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shousheng Xu
- School of Sports Engineering, Beijing Sport University, Beijing, China
| |
Collapse
|
94
|
De Giovanni M, Dang EV, Chen KY, An J, Madhani HD, Cyster JG. Platelets and mast cells promote pathogenic eosinophil recruitment during invasive fungal infection via the 5-HIAA-GPR35 ligand-receptor system. Immunity 2023; 56:1548-1560.e5. [PMID: 37279752 PMCID: PMC10360074 DOI: 10.1016/j.immuni.2023.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Cryptococcus neoformans is the leading cause of fungal meningitis and is characterized by pathogenic eosinophil accumulation in the context of type-2 inflammation. The chemoattractant receptor GPR35 is expressed by granulocytes and promotes their migration to the inflammatory mediator 5-hydroxyindoleacetic acid (5-HIAA), a serotonin metabolite. Given the inflammatory nature of cryptococcal infection, we examined the role of GPR35 in the circuitry underlying cell recruitment to the lung. GPR35 deficiency dampened eosinophil recruitment and fungal growth, whereas overexpression promoted eosinophil homing to airways and fungal replication. Activated platelets and mast cells were the sources of GPR35 ligand activity and pharmacological inhibition of serotonin conversion to 5-HIAA, or genetic deficiency in 5-HIAA production by platelets and mast cells resulted in more efficient clearance of Cryptococcus. Thus, the 5-HIAA-GPR35 axis is an eosinophil chemoattractant receptor system that modulates the clearance of a lethal fungal pathogen, with implications for the use of serotonin metabolism inhibitors in the treatment of fungal infections.
Collapse
Affiliation(s)
- Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Eric V Dang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin Y Chen
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hiten D Madhani
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
95
|
Matera MG, Rogliani P, Ora J, Calzetta L, Cazzola M. A comprehensive overview of investigational elastase inhibitors for the treatment of acute respiratory distress syndrome. Expert Opin Investig Drugs 2023; 32:793-802. [PMID: 37740909 DOI: 10.1080/13543784.2023.2263366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 09/22/2023] [Indexed: 09/25/2023]
Abstract
INTRODUCTION Excessive activity of neutrophil elastase (NE), the main enzyme present in azurophil granules in the neutrophil cytoplasm, may cause tissue injury and remodeling in various lung diseases, including acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), in which it is crucial to the immune response and inflammatory process. Consequently, NE is a possible target for therapeutic intervention in ALI/ARDS. AREAS COVERED The protective effects of several NE inhibitors in attenuating ALI/ARDS in several models of lung injury are described. Some of these NE inhibitors are currently in clinical development, but only sivelestat has been evaluated as a treatment for ALI/ARDS. EXPERT OPINION Preclinical research has produced encouraging information about using NE inhibitors. Nevertheless, only sivelestat has been approved for this clinical indication, and only in Japan and South Korea because of the conflicting results of clinical trials and likely also because of the potential adverse events. Identifying subsets of patients with ARDS most likely to benefit from NE inhibitor treatment, such as the hyperinflammatory phenotype, and using a more advanced generation of NE inhibitors than sivelestat could enable better clinical results than those obtained with elastase inhibitors.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Disease and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
96
|
Arora A, Singh A. Exploring the role of neutrophils in infectious and noninfectious pulmonary disorders. Int Rev Immunol 2023; 43:41-61. [PMID: 37353973 DOI: 10.1080/08830185.2023.2222769] [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: 03/25/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
With the change in global environment, respiratory disorders are becoming more threatening to the health of people all over the world. These diseases are closely linked to performance of immune system. Within the innate arm of immune system, Neutrophils are an important moiety to serve as an immune defense barrier. They are one of the first cells recruited to the site of infection and plays a critical role in pathogenesis of various pulmonary diseases. It is established that the migration and activation of neutrophils can lead to inflammation either directly or indirectly and this inflammation caused is very crucial for the clearance of pathogens and resolution of infection. However, the immunopathological mechanisms involved to carry out the same is very complex and not well understood. Despite there being studies concentrating on the role of neutrophils in multiple respiratory diseases, there is still a long way to go in order to completely understand the complexity of the participation of neutrophils and mechanisms involved in the development of these respiratory diseases. In the present article, we have reviewed the literature to comprehensively provide an insight in the current development and advancements about the role of neutrophils in infectious respiratory disorders including viral respiratory disorders such as Coronavirus disease (COVID-19) and bacterial pulmonary disorders with a focused review on pulmonary tuberculosis as well as in noninfectious disorders like Chronic obstructive pulmonary disease (COPD) and asthma. Also, future directions into research and therapeutic targets have been discussed for further exploration.
Collapse
Affiliation(s)
- Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
97
|
Han F, Ding ZF, Shi XL, Zhu QT, Shen QH, Xu XM, Zhang JX, Gong WJ, Xiao WM, Wang D, Chen WW, Hu LH, Lu GT. Irisin inhibits neutrophil extracellular traps formation and protects against acute pancreatitis in mice. Redox Biol 2023; 64:102787. [PMID: 37392517 DOI: 10.1016/j.redox.2023.102787] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 07/03/2023] Open
Abstract
INTRODUCTION Irisin is a newly discovered myokine which links exercise to inflammation and inflammation-related diseases through macrophage regulation. However, the effect of irisin on the activity of inflammation related immune cells (such as neutrophils) has not been clearly described. OBJECTIVES The objective of our study was to explore the effect of irisin on the neutrophil extracellular traps (NETs) formation. METHODS Phorbol-12-myristate-13-acetate (PMA) was used to construct a classic neutrophil inflammation model that was used to observe the formation of NETs in vitro. We studied the effect of irisin on NETs formation and its regulation mechanism. Subsequently, acute pancreatitis (AP) was used to verify the protective effect of irisin in vivo, which was an acute aseptic inflammatory response disease model closely related to NETs. RESULTS Our study found that addition of irisin significantly reduced the formation of NETs via regulation of the P38/MAPK pathway through integrin αVβ5, which might be the one of key pathways in NETs formation, and which could theoretically offset the immunoregulatory effect of irisin. Systemic treatment with irisin reduced the severity of tissue damage common in the disease and inhibited the formation of NETs in pancreatic necrotic tissue of two classical AP mouse models. CONCLUSION The findings confirmed for the first time that irisin could inhibit NETs formation and protect mice from pancreatic injury, which further elucidated the protective effect of exercise on acute inflammatory injury.
Collapse
Affiliation(s)
- Fei Han
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zi-Fan Ding
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; International Sport Management, Health and Life Sciences, Northumbria University Newcastle, NE1 8ST, UK
| | - Xiao-Lei Shi
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qing-Tian Zhu
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qin-Hao Shen
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xing-Meng Xu
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun-Xian Zhang
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei-Juan Gong
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei-Ming Xiao
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Dan Wang
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Naval Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wei-Wei Chen
- Department of Gastroenterology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Liang-Hao Hu
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Naval Medical University, Shanghai, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Guo-Tao Lu
- Pancreatic Center, Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China; Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
98
|
Lacey KA, Serpas L, Makita S, Wang Y, Rashidfarrokhi A, Soni C, Gonzalez S, Moreira A, Torres VJ, Reizis B. Secreted mammalian DNases protect against systemic bacterial infection by digesting biofilms. J Exp Med 2023; 220:e20221086. [PMID: 36928522 PMCID: PMC10037111 DOI: 10.1084/jem.20221086] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
Extracellular DNase DNASE1L3 maintains tolerance to self-DNA in humans and mice, whereas the role of its homolog DNASE1 remains controversial, and the overall function of secreted DNases in immunity is unclear. We report that deletion of murine DNASE1 neither caused autoreactivity in isolation nor exacerbated lupus-like disease in DNASE1L3-deficient mice. However, combined deficiency of DNASE1 and DNASE1L3 rendered mice susceptible to bloodstream infection with Staphylococcus aureus. DNASE1/DNASE1L3 double-deficient mice mounted a normal innate response to S. aureus and did not accumulate neutrophil extracellular traps (NETs). However, their kidneys manifested severe pathology, increased bacterial burden, and biofilm-like bacterial lesions that contained bacterial DNA and excluded neutrophils. Furthermore, systemic administration of recombinant DNASE1 protein during S. aureus infection rescued the mortality of DNase-deficient mice and ameliorated the disease in wild-type mice. Thus, DNASE1 and DNASE1L3 jointly facilitate the control of bacterial infection by digesting extracellular microbial DNA in biofilms, suggesting the original evolutionary function of secreted DNases as antimicrobial agents.
Collapse
Affiliation(s)
- Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lee Serpas
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sohei Makita
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yueyang Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Andre Moreira
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
99
|
Sejópoles MD, Souza-Silva JP, Silva-Santos C, Paula-Duarte MM, Fontes CJ, Gomes LT. Prognostic value of neutrophil and lymphocyte counts and neutrophil/lymphocyte ratio for predicting death in patients hospitalized for COVID-19. Heliyon 2023; 9:e16964. [PMID: 37292322 PMCID: PMC10238120 DOI: 10.1016/j.heliyon.2023.e16964] [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: 10/26/2022] [Revised: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
COVID-19 primarily affects the respiratory system and can cause changes in other systems. Early identification of patients with a higher potential for complications is critical to provide the best possible treatment to reduce the disease's lethality. This study aimed to analyze the behavior of hematologic biomarkers in predicting mortality in patients hospitalized with COVID-19. This retrospective cohort study used data from the medical records of patients hospitalized with COVID-19 between March and August 2020 in two referral hospitals for treatment of the disease in the city of Cuiabá (in the state of Mato Grosso, Brazil). Clinical and laboratory characteristics related to cardiovascular involvement and death during hospitalization were evaluated. Neutrophils, lymphocytes, and monocytes, as well as the neutrophil-to-lymphocyte ratio (NLR) and the monocyte-to-lymphocyte ratio (MRL), were used as potential biomarkers of death. A total of 199 patients were included (male: 113; mean age: 51.4 years). Leukocyte, neutrophil, and lymphocyte counts showed a statistically significant association with death, as did NLR and MRL. Satisfactory accuracy in predicting death was observed for leukocyte, neutrophil, lymphocyte, NLR, and MLR counts. The hematologic biomarkers studied may be useful for prognosticating hospitalized patients for the possibility of death from COVID-19.
Collapse
Affiliation(s)
- Marcia D Sejópoles
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
| | - João P Souza-Silva
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
- Faculty of Medicine. Post-Graduation in Health Sciences, Federal University of Mato Grosso, Av Fernando Correa da Costa, 2367, Cuiabá (MT), Brazil
| | - Cristiane Silva-Santos
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
- Faculty of Medicine. Post-Graduation in Health Sciences, Federal University of Mato Grosso, Av Fernando Correa da Costa, 2367, Cuiabá (MT), Brazil
| | - Matheus M Paula-Duarte
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
- Faculty of Medicine. Post-Graduation in Health Sciences, Federal University of Mato Grosso, Av Fernando Correa da Costa, 2367, Cuiabá (MT), Brazil
| | - Cor Jf Fontes
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
- Faculty of Medicine. Post-Graduation in Health Sciences, Federal University of Mato Grosso, Av Fernando Correa da Costa, 2367, Cuiabá (MT), Brazil
| | - Luciano T Gomes
- Julio Müller School Hospital, Federal University of Mato Grosso. Rua Dr Luis P Pereira Leite Sn, Cuiabá (MT), Brazil
- Faculty of Medicine. Post-Graduation in Health Sciences, Federal University of Mato Grosso, Av Fernando Correa da Costa, 2367, Cuiabá (MT), Brazil
| |
Collapse
|
100
|
Zhu Y, Luo Y, Li L, Jiang X, Du Y, Wang J, Li H, Gu H, Li D, Tang H, Qin H, Xu C, Liu Y, Zhao D, Guo Y, Liu F. Immune response plays a role in Mycoplasma pneumoniae pneumonia. Front Immunol 2023; 14:1189647. [PMID: 37304280 PMCID: PMC10250694 DOI: 10.3389/fimmu.2023.1189647] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Mycoplasma pneumoniae (MP) is a major pathogen of community-acquired pneumonia in children. However, the specific pathogenesis of the progression of Mycoplasma pneumoniae pneumonia (MPP) is unclear. We aimed to reveal the landscape of microbiota and the host immune response in MPP. Methods This self-controlled study analyzed the microbiome and transcriptome of bronchoalveolar lavage fluid (BALF) from the severe side (SD) and opposite side (OD) of 41 children with MPP from January to December 2021 and revealed the differences of the peripheral blood neutrophil function among children with mild MPP, severe MPP, and healthy children through transcriptome sequencing. Results The MP load or the pulmonary microbiota had no significant difference between the SD group and OD group, and the deterioration of MPP was related to the immune response, especially the intrinsic immune response. Discussion The immune response plays a role in MPP, which may inform treatment strategies for MPP.
Collapse
Affiliation(s)
- Yifan Zhu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yingying Luo
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Li
- Department of Respiratory Medicine, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xinyi Jiang
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Du
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Wang
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Huilin Li
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyan Gu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Daiying Li
- Vision Medicals Center for Infectious Diseases, Guangzhou, China
| | - Heng Tang
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Houbing Qin
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Changdi Xu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Liu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Guo
- Department of Respiratory Medicine, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Feng Liu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|