Analgesics
Antiandrogens
Azvudine
Bromhexine
Budesonide
Colchicine
Conv. Plasma
Curcumin
Famotidine
Favipiravir
Fluvoxamine
Hydroxychlor..
Ivermectin
Lifestyle
Melatonin
Metformin
Minerals
Molnupiravir
Monoclonals
Naso/orophar..
Nigella Sativa
Nitazoxanide
Paxlovid
Quercetin
Remdesivir
Thermotherapy
Vitamins
More

Other
Feedback
Home
Top
Abstract
All ivermectin studies
Meta analysis
 
Feedback
Home
next
study
previous
study
c19ivm.org COVID-19 treatment researchIvermectinIvermectin (more..)
Melatonin Meta
Metformin Meta
Azvudine Meta
Bromhexine Meta Molnupiravir Meta
Budesonide Meta
Colchicine Meta
Conv. Plasma Meta Nigella Sativa Meta
Curcumin Meta Nitazoxanide Meta
Famotidine Meta Paxlovid Meta
Favipiravir Meta Quercetin Meta
Fluvoxamine Meta Remdesivir Meta
Hydroxychlor.. Meta Thermotherapy Meta
Ivermectin Meta

All Studies   Meta Analysis    Recent:   

Crosstalk between neutrophil extracellular traps and immune regulation: insights into pathobiology and therapeutic implications of transfusion-related acute lung injury

Liu et al., Frontiers in Immunology, doi:10.3389/fimmu.2023.1324021
Dec 2023  
  Post
  Facebook
Share
  Source   PDF   All Studies   Meta AnalysisMeta
Ivermectin for COVID-19
4th treatment shown to reduce risk in August 2020
 
*, now known with p < 0.00000000001 from 102 studies, recognized in 22 countries.
No treatment is 100% effective. Protocols combine complementary and synergistic treatments. * >10% efficacy in meta analysis with ≥3 clinical studies.
4,100+ studies for 60+ treatments. c19ivm.org
Ivermectin may be beneficial for COVID-19 ARDS by blocking GSDMD and NET formation.
Authors review the role of neutrophil extracellular traps (NETs) in transfusion-related acute lung injury (TRALI). Authors discusses the mechanisms of NET formation, including vital NETosis and NETosis involving cell death. They examine the evidence showing that NETs contribute to endothelial and lung epithelial damage in TRALI. Authors explore the interactions of NETs with other immune cells like macrophages, dendritic cells, and T cells, which create inflammatory feedback loops that exacerbate tissue injury. Authors suggest potential therapeutic approaches targeting NET formation, NET clearance, cytokine signaling, and glucose metabolism pathways to dampen detrimental inflammation in TRALI.
Authors note that ivermectin was shown to inhibit GSDMD oligomerization, alleviating the release of NETs, and that increased NET formation was observed in ARDS induced by COVID-19.
This suggests that ivermectin may be beneficial for COVID-19 ARDS by inhibiting NET formation. Specifically: ivermectin can inhibit GSDMD oligomerization, which the authors note plays an important role in NET release. By blocking GSDMD, ivermectin may suppress detrimental NETosis. Authors state that in COVID-19 ARDS, there is downregulation of respiratory bursts but increased NET formation. So while ROS production from neutrophils may be impaired, excessive NETs are still an issue driving lung damage. By alleviating NET release through GSDMD inhibition, ivermectin could therefore target one of the key pathological pathways - NETosis - that is overactivated in COVID-19 ARDS.
Ivermectin, better known for antiparasitic activity, is a broad spectrum antiviral with activity against many viruses including H7N7 Götz, Dengue Jitobaom, Tay, Wagstaff, HIV-1 Wagstaff, Simian virus 40 Wagstaff (B), Zika Barrows, Jitobaom, Yang, West Nile Yang, Yellow Fever Mastrangelo, Varghese, Japanese encephalitis Mastrangelo, Chikungunya Varghese, Semliki Forest virus Varghese, Human papillomavirus Li, Epstein-Barr Li, BK Polyomavirus Bennett, and Sindbis virus Varghese.
Ivermectin inhibits importin-α/β-dependent nuclear import of viral proteins Götz, Kosyna, Wagstaff, Wagstaff (B), shows spike-ACE2 disruption at 1nM with microfluidic diffusional sizing Fauquet, binds to glycan sites on the SARS-CoV-2 spike protein preventing interaction with blood and epithelial cells and inhibiting hemagglutination Boschi, Scheim, shows dose-dependent inhibition of wildtype and omicron variants Shahin, exhibits dose-dependent inhibition of lung injury Abd-Elmawla, Ma, may inhibit SARS-CoV-2 via IMPase inhibition Jitobaom, may inhibit SARS-CoV-2 induced formation of fibrin clots resistant to degradation Vottero, inhibits SARS-CoV-2 3CLpro Mody, may inhibit SARS-CoV-2 RdRp activity Parvez (B), may minimize viral myocarditis by inhibiting NF-κB/p65-mediated inflammation in macrophages Gao, may be beneficial for COVID-19 ARDS by blocking GSDMD and NET formation Liu (C), shows protection against inflammation, cytokine storm, and mortality in an LPS mouse model sharing key pathological features of severe COVID-19 DiNicolantonio, Zhang, may be beneficial in severe COVID-19 by binding IGF1 to inhibit the promotion of inflammation, fibrosis, and cell proliferation that leads to lung damage Zhao, may minimize SARS-CoV-2 induced cardiac damage Liu, Liu (B), increases Bifidobacteria which play a key role in the immune system Hazan, has immunomodulatory Munson and anti-inflammatory DiNicolantonio (B), Yan properties, and has an extensive and very positive safety profile Descotes.
Liu et al., 7 Dec 2023, peer-reviewed, 9 authors.
This PaperIvermectinAll
Crosstalk between neutrophil extracellular traps and immune regulation: insights into pathobiology and therapeutic implications of transfusion-related acute lung injury
Yi Liu, Rong Wang, Congkuan Song, Song Ding, Yifan Zuo, Ke Yi, Ning Li, Bo Wang, Qing Geng
Frontiers in Immunology, doi:10.3389/fimmu.2023.1324021
Transfusion-related acute lung injury (TRALI) is the leading cause of transfusionassociated death, occurring during or within 6 hours after transfusion. Reports indicate that TRALI can be categorized as having or lacking acute respiratory distress syndrome (ARDS) risk factors. There are two types of TRALI in terms of its pathogenesis: antibody-mediated and non-antibody-mediated. The key initiation steps involve the priming and activation of neutrophils, with neutrophil extracellular traps (NETs) being established as effector molecules formed by activated neutrophils in response to various stimuli. These NETs contribute to the production and release of reactive oxygen species (ROS) and participate in the destruction of pulmonary vascular endothelial cells. The significant role of NETs in TRALI is well recognized, offering a potential pathway for TRALI treatment. Moreover, platelets, macrophages, endothelial cells, and complements have been identified as promoters of NET formation. Concurrently, studies have demonstrated that the storage of platelets and concentrated red blood cells (RBC) can induce TRALI through bioactive lipids. In this article, recent clinical and pre-clinical studies on the pathophysiology and pathogenesis of TRALI are reviewed to further illuminate the mechanism through which NETs induce TRALI. This review aims to propose new therapeutic strategies for TRALI, with the hope of effectively improving its poor prognosis.
Conflict of interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. Publisher's note All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher. Appendix The proteins/genes described in this review.
References
Adrover, Carrau, Daßler-Plenker, Bram, Chandar et al., Disulfiram inhibits neutrophil extracellular trap formation and protects rodents from acute lung injury and SARS-CoV-2 infection, JCI Insight, doi:10.1172/jci.insight.157342
Alfaro, Teijeira, Onate, Perez, Sanmamed et al., Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs), Clin Cancer Res, doi:10.1158/1078-0432.CCR-15-2463
Amara, Cooper, Voronkova, Webb, Lynch et al., Selective activation of PFKL suppresses the phagocytic oxidative burst, Cell, doi:10.1016/j.cell.2021.07.004
Apel, Andreeva, Knackstedt, Streeck, Frese et al., The cytosolic DNA sensor cGAS recognizes neutrophil extracellular traps, Sci Signal, doi:10.1126/scisignal.aax7942
Awasthi, Nagarkoti, Sadaf, Chandra, Kumar et al., Glycolysis dependent lactate formation in neutrophils: A metabolic link between NOX-dependent and independent NETosis, Biochim Biophys Acta Mol Basis Dis, doi:10.1016/j.bbadis.2019.165542
Aymonnier, Ng, Fredenburgh, Zambrano-Vera, Munzer et al., Inflammasome activation in neutrophils of patients with severe COVID-19, Blood Adv, doi:10.1182/bloodadvances.2021005949
Azevedo, Rochael, Guimaraes-Costa, De Souza-Vieira, Ganilho et al., A metabolic shift toward pentose phosphate pathway is necessary for amyloid fibriland phorbol 12-myristate 13-acetate-induced neutrophil extracellular trap (NET) formation, J Biol Chem, doi:10.1074/jbc.M115.640094
Baudel, Vigneron, Pras-Landre, Joffre, Marjot et al., Transfusion-related acute lung injury (TRALI) after intravenous immunoglobulins: French multicentre study and literature review, Clin Rheumatol, doi:10.1007/s10067-019-04832-7
Bertini, Allegretti, Bizzarri, Moriconi, Locati et al., Noncompetitive allosteric inhibitors of the inflammatory chemokine receptors CXCR1 and CXCR2: prevention of reperfusion injury, Proc Natl Acad Sci, doi:10.1084/jem.20181102
Borella, Biasi, Paolini, Boraldi, Tartaro et al., Metabolic reprograming shapes neutrophil functions in severe COVID-19, Eur J Immunol, doi:10.1002/eji.202149481
Brinkmann, Reichard, Goosmann, Fauler, Uhlemann et al., Neutrophil extracellular traps kill bacteria, Science, doi:10.1126/science.1092385
Britt, Lika, Giese, Schoen, Seim et al., Switching to the cyclic pentose phosphate pathway powers the oxidative burst in activated neutrophils, Nat Metab, doi:10.1038/s42255-022-00550-8
Cao, Zhang, Wang, Yang, Ma et al., Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy, Proc Natl Acad Sci, doi:10.1016/j.ccell.2016.10.006
Caudrillier, Kessenbrock, Gilliss, Nguyen, Marques et al., Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury, J Clin Invest, doi:10.1172/JCI61303
Chapman, Lyon, Simpson, Mason, Beynon et al., Caught in a trap? Proteomic analysis of neutrophil extracellular traps in rheumatoid arthritis and systemic lupus erythematosus, Front Immunol, doi:10.3389/fimmu.2019.00423
Chauhan, Demon, Walle, Paerewijck, Zecchin et al., GSDMD drives canonical inflammasome-induced neutrophil pyroptosis and is dispensable for NETosis, EMBO Rep, doi:10.15252/embr.202154277
Chen, Gross, Sotomayor, Stacey, Tschopp et al., The neutrophil NLRC4 inflammasome selectively promotes IL-1beta maturation without pyroptosis during acute Salmonella challenge, Cell Rep, doi:10.1016/j.celrep.2014.06.028
Chen, Monteleone, Boucher, Sollberger, Ramnath et al., Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps, Sci Immunol, doi:10.1126/sciimmunol.aar6676
Chen, Zhao, Lai, Zhang, Yang et al., Neutrophil extracellular traps promote macrophage pyroptosis in sepsis, Cell Death Dis, doi:10.1038/s41419-017-0090-8
Cieloch, Kuzmicka, Mroczek, Stelmaszczyk-Emmel, Demkow, Secretomes of M1 and M2 macrophages decrease the release of neutrophil extracellular traps, Sci Rep, doi:10.1038/s41598-023-42167-1
Cleary, Kwaan, Tian, Calabrese, Mallavia et al., C5aR1 signaling triggers lung immunopathology in COVID-19 through neutrophil extracellular traps, J Clin Invest, doi:10.1172/JCI163105
Cui, Yang, Tao, Peng, Luo et al., Neutrophil extracellular traps induce alveolar macrophage pyroptosis by regulating NLRP3 deubiquitination, aggravating the development of septic lung injury, J Inflamm Res, doi:10.2147/JIR.S366436
Domer, Walther, Moller, Behnen, Laskay, Neutrophil extracellular traps activate proinflammatory functions of human neutrophils, Front Immunol, doi:10.3389/fimmu.2021.636954
Doring, Libby, Soehnlein, Neutrophil extracellular traps participate in cardiovascular diseases: recent experimental and clinical insights, Circ Res, doi:10.1161/CIRCRESAHA.120.315931
Douda, Khan, Grasemann, Palaniyar, SK3 channel and mitochondrial ROS mediate NADPH oxidase-independent NETosis induced by calcium influx, Proc Natl Acad Sci, doi:10.1073/pnas.1414055112
Eelen, De Zeeuw, Simons, Carmeliet, Endothelial cell metabolism in normal and diseased vasculature, Circ Res, doi:10.1161/CIRCRESAHA.116.302855
Farrera, Fadeel, Macrophage clearance of neutrophil extracellular traps is a silent process, J Immunol, doi:10.4049/jimmunol.1300436
Folco, Mawson, Vromman, Bernardes-Souza, Franck et al., Neutrophil extracellular traps induce endothelial cell activation and tissue factor production through interleukin-1alpha and cathepsin G, Arterioscler Thromb Vasc Biol, doi:10.1161/ATVBAHA.118.311150
Ghergurovich, Garcia-Canaveras, Wang, Schmidt, Zhang et al., A small molecule G6PD inhibitor reveals immune dependence on pentose phosphate pathway, Nat Chem Biol, doi:10.1038/s41589-020-0533-x
Goldspink, Schmitz, Babyak, Brauns, Milleck et al., Kidney medullary sodium chloride concentrations induce neutrophil and monocyte extracellular DNA traps that defend against pyelonephritis, vivo Kidney Int, doi:10.1016/j.kint.2023.03.034
Gomes, Varady, Lourenco, Mizurini, Rondon et al., IL-1beta blockade attenuates thrombosis in a neutrophil extracellular trap-dependent breast cancer model, Front Immunol, doi:10.3389/fimmu.2019.02088
Gregoire, Uhel, Lesouhaitier, Gacouin, Guirriec et al., Impaired efferocytosis and neutrophil extracellular trap clearance by macrophages in ARDS, Eur Respir J, doi:10.1183/13993003.02590-2017
Haider, Kral-Pointner, Mayer, Richter, Kaun et al., Neutrophil extracellular trap degradation by differently polarized macrophage subsets, Arterioscler Thromb Vasc Biol, doi:10.1161/ATVBAHA.120.314883
Herre, Cedervall, Mackman, Olsson, Neutrophil extracellular traps in the pathology of cancer and other inflammatory diseases, Physiol Rev, doi:10.1152/physrev.00062.2021
Hu, Liu, Xia, Zhang, Zhang et al., FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation, Nat Immunol, doi:10.1038/s41590-020-0669-6
Humphries, Shmuel-Galia, Ketelut-Carneiro, Li, Wang et al., Necrostatin-1 alleviates diffuse pulmonary haemorrhage by preventing the release of NETs via inhibiting NE/ GSDMD activation in murine lupus, Front Immunol, doi:10.1155/2023/4743975
Jaillon, Peri, Delneste, Fremaux, Doni et al., The humoral pattern recognition receptor PTX3 is stored in neutrophil granules and localizes in extracellular traps, J Exp Med, doi:10.1084/jem.20061301
Jayne, Merkel, Schall, Bekker, Group, Avacopan for the treatment of ANCA-associated vasculitis, N Engl J Med, doi:10.1056/NEJMoa2023386
Jiao, Zhang, Liu, Zhou, Qi et al., Exosomal PGE2 from M2 macrophages inhibits neutrophil recruitment and NET formation through lipid mediator class switching in sepsis, J BioMed Sci, doi:10.1186/s12929-023-00957-9
Kambara, Liu, Zhang, Liu, Bajrami et al., Gasdermin D exerts anti-inflammatory effects by promoting neutrophil death, Cell Rep, doi:10.1016/j.celrep.2018.02.067
Kapur, Kasetty, Rebetz, Egesten, Semple et al., Osteopontin mediates murine transfusion-related acute lung injury via stimulation of pulmonary neutrophil accumulation, Nature, doi:10.1038/s41586-022-04802-1
Karmakar, Minns, Greenberg, Diaz-Aponte, Pestonjamasp et al., N-GSDMD trafficking to neutrophil organelles facilitates IL-1beta release independently of plasma membrane pores and pyroptosis, Nat Commun, doi:10.1038/s41467-020-16043-9
Kim, Jang, Dharaneeswaran, Li, Bhide et al., Endothelial pyruvate kinase M2 maintains vascular integrity, J Clin Invest, doi:10.1172/JCI120912
Kono, Saigo, Takagi, Kawauchi, Wada et al., Morphological and flow-cytometric analysis of haemin-induced human neutrophil activation: implications for transfusion-related acute lung injury, Blood Transfus, doi:10.2450/2012.0141-11
Kono, Saigo, Takagi, Takahashi, Kawauchi et al., Hemerelated molecules induce rapid production of neutrophil extracellular traps, Transfusion, doi:10.1111/trf.12700
Kovacs, Oh, Maltez, Mcglaughon, Verma et al., Neutrophil caspase-11 is essential to defend against a cytosol-invasive bacterium, Cell Rep, doi:10.1016/j.celrep.2020.107967
Lambeth, Neish, Nox enzymes and new thinking on reactive oxygen: a double-edged sword revisited, Annu Rev Pathol, doi:10.1146/annurev-pathol-012513-104651
Lande, Ganguly, Facchinetti, Frasca, Conrad et al., Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus, Sci Transl Med, doi:10.1126/scitranslmed.3001180
Lande, Gregorio, Facchinetti, Chatterjee, Wang et al., Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide, Nature, doi:10.1038/nature06116
Lauth, Kockritz-Blickwede, Mcnamara, Myskowski, Zinkernagel et al., M1 protein allows Group A streptococcal survival in phagocyte extracellular traps through cathelicidin inhibition, J Innate Immun, doi:10.1159/000203645
Lazzaretto, Fadeel, Intra-and extracellular degradation of neutrophil extracellular traps by macrophages and dendritic cells, J Immunol, doi:10.4049/jimmunol.1800159
Le, Wu, Liu, Wu, Hu et al., MiR-144-induced KLF2 inhibition and NF-kappaB/CXCR1 activation promote neutrophil extracellular trap-induced transfusion-related acute lung injury, J Cell Mol Med, doi:10.1111/jcmm.16650
Leffler, Martin, Gullstrand, Tyden, Lood et al., Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease, J Immunol, doi:10.4049/jimmunol.1102404
Leppkes, Knopf, Naschberger, Lindemann, Singh et al., Vascular occlusion by neutrophil extracellular traps in COVID-19, EBioMedicine, doi:10.1016/j.ebiom.2020.102925
Li, Hook, Ding, Chung, Mettlen, Neutrophil metabolomics in severe COVID-19 reveal GAPDH as a suppressor of neutrophil extracellular trap formation, Nat Commun, doi:10.1038/s41467-023-37567-w
Li, Wang, Liu, Li, Zhang et al., Neutrophil extracellular traps induce intestinal damage and thrombotic tendency in inflammatory bowel disease, J Crohns Colitis, doi:10.1093/ecco-jcc/jjz132
Lin, Hu, Hu, Li, Wang et al., NET-triggered NLRP3 activation and IL18 release drive oxaliplatin-induced peripheral neuropathy, Cancer Immunol Res, doi:10.1158/2326-6066.CIR-22-0197
Liu, Xiao, Zhou, Wang, Peng et al., PFKFB3 promotes sepsisinduced acute lung injury by enhancing NET formation by CXCR4(hi) neutrophils, Int Immunopharmacol, doi:10.1016/j.intimp.2023.110737
Liu, Yang, Gao, Yu, Shi et al., NLRP3 activation induced by neutrophil extracellular traps sustains inflammatory response in the diabetic wound, Clin Sci (Lond), doi:10.1042/CS20180600
Lood, Blanco, Purmalek, Carmona-Rivera, Ravin et al., Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease, Nat Med, doi:10.1038/nm.4027
Luo, Dai, Feng, Ding, Shao et al., Suppression of lncRNA NLRP3 inhibits NLRP3-triggered inflammatory responses in early acute lung injury, Cell Death Dis, doi:10.1038/s41419-021-04180-y
Mcdonald, Davis, Kim, Tse, Esmon et al., Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice, Blood, doi:10.1182/blood-2016-09-741298
Metzler, Goosmann, Lubojemska, Zychlinsky, Papayannopoulos, A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis, Cell Rep, doi:10.1016/j.celrep.2014.06.044
Mistry, Nakabo, Neil, Goel, Jiang et al., Transcriptomic, epigenetic, and functional analyses implicate neutrophil diversity in the pathogenesis of systemic lupus erythematosus, Proc Natl Acad Sci, doi:10.1073/pnas.1908576116
Munzer, Negro, Fukui, Di Meglio, Aymonnier et al., NLRP3 inflammasome assembly in neutrophils is supported by PAD4 and promotes NETosis under sterile conditions, Front Immunol, doi:10.3389/fimmu.2021.683803
Papayannopoulos, Neutrophil extracellular traps in immunity and disease, Nat Rev Immunol, doi:10.1038/nri.2017.105
Peters, Van Hezel, Cortjens, Boer, Van Bruggen et al., Transfusion of 35-day stored RBCs in the presence of endotoxemia does not result in lung injury in humans, Crit Care Med, doi:10.1097/CCM.0000000000001614
Peters, Van Stein, Vlaar, Antibody-mediated transfusion-related acute lung injury; from discovery to prevention, Br J Haematol, doi:10.1111/bjh.13459
Peters, Vervaart, Van Bruggen, De Korte, Nieuwland et al., Non-polar lipids accumulate during storage of transfusion products and do not 12, Immunity, doi:10.1016/j.immuni.2021.06.006
Pilsczek, Salina, Poon, Fahey, Yipp et al., A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus, J Immunol, doi:10.4049/jimmunol.1000675
Rathkey, Zhao, Liu, Chen, Yang et al., Chemical disruption of the pyroptotic pore-forming protein gasdermin D inhibits inflammatory cell death and sepsis, Sci Immunol, doi:10.1126/sciimmunol.aat2738
Rodriguez-Espinosa, Rojas-Espinosa, Moreno-Altamirano, Lopez-Villegas, Sanchez-Garcia, Metabolic requirements for neutrophil extracellular traps formation, Immunology, doi:10.1111/imm.12437
Sadiku, Willson, Dickinson, Murphy, Harris et al., Prolyl hydroxylase 2 inactivation enhances glycogen storage and promotes excessive neutrophilic responses, J Clin Invest, doi:10.1172/JCI90848
Sano, Maejima, Nakagama, Shiheido-Watanabe, Tamura et al., Neutrophil extracellular traps-mediated Beclin-1 suppression aggravates atherosclerosis by inhibiting macrophage autophagy, Front Cell Dev Biol, doi:10.3389/fcell.2022.876147
Santoni, Pericat, Gorse, Buyck, Pinilla et al., Caspase-1driven neutrophil pyroptosis and its role in host susceptibility to Pseudomonas aeruginosa, PloS Pathog, doi:10.1371/journal.ppat.1010305
Schauer, Janko, Munoz, Zhao, Kienhofer et al., Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines, Nat Med, doi:10.1038/nm.3547
Schmickl, Mastrobuoni, Filippidis, Shah, Radic et al., Male-predominant plasma transfusion strategy for preventing transfusion-related acute lung injury: a systematic review, Crit Care Med, doi:10.1097/CCM.0000000000000675
Schneider, Machado, Veras, Maganin, De Souza et al., Neutrophil extracellular traps mediate joint hyperalgesia induced by immune inflammation, Rheumatol, doi:10.1093/rheumatology/keaa794
Schnitzler, Hoogeveen, Ali, Prange, Waissi et al., Atherogenic lipoprotein(a) increases vascular glycolysis, thereby facilitating inflammation and leukocyte extravasation, Circ Res, doi:10.1161/CIRCRESAHA.119.316206
Semple, Rebetz, Kapur, Transfusion-associated circulatory overload and transfusion-related acute lung injury, Blood, doi:10.1182/blood-2018-10-860809
Shi, Liu, Cao, Ma, Zhu et al., Inhibition of neutrophil extracellular trap formation ameliorates neuroinflammation and neuronal apoptosis via STING-dependent IRE1alpha/ASK1/JNK signaling pathway in mice with traumatic brain injury, J Neuroinflamm, doi:10.1186/s12974-023-02903-w
Shi, Zhao, Wang, Shi, Wang et al., Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death, Nature, doi:10.1038/nature15514
Silliman, Kelher, Khan, Lasarre, West et al., Experimental prestorage filtration removes antibodies and decreases lipids in RBC supernatants mitigating TRALI in vivo, Blood, doi:10.1182/blood-2013-10-532424
Silliman, The two-event model of transfusion-related acute lung injury, Crit Care Med, doi:10.1097/01.CCM.0000214292.62276.8E
Silva, Wanderley, Veras, Sonego, Nascimento et al., Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation, Crit Care, doi:10.1186/s13054-022-04062-5
Skov, Beurskens, Zachariae, Reitamo, Teeling et al., IL-8 as antibody therapeutic target in inflammatory diseases: reduction of clinical activity in palmoplantar pustulosis, J Immunol, doi:10.4049/jimmunol.181.1.669
Sollberger, Choidas, Burn, Habenberger, Lucrezia et al., Gasdermin D plays a vital role in the generation of neutrophil extracellular traps, Sci Immunol, doi:10.1126/sciimmunol.aar6689
Stojkov, Claus, Kozlowski, Oberson, Scharen et al., NET formation is independent of gasdermin D and pyroptotic cell death, Sci Signal, doi:10.1126/scisignal.abm0517
Suksawad, Udompornpitak, Thawinpipat, Korwattanamongkol, Visitchanakun et al., Cyclic GMP-AMP synthase (cGAS) deletion reduces severity in bilateral nephrectomy mice through changes in neutrophil extracellular traps and mitochondrial respiration, Biomedicines, doi:10.3390/biomedicines11041208
Swiecki, Colonna, The multifaceted biology of plasmacytoid dendritic cells, Nat Rev Immunol, doi:10.1038/nri3865
Teijeira, Garasa, Gato, Alfaro, Migueliz et al., CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity, Immunity, doi:10.1016/j.immuni.2020.03.001
Thomas, Carbo, Curtis, Martinod, Mazo et al., Extracellular DNA traps are associated with the pathogenesis of TRALI in humans and mice, Blood, doi:10.1182/blood-2012-01-405183
Torres Caban, Yang, Lai, Yang, Subach et al., Tuning the sensitivity of genetically encoded fluorescent potassium indicators through structure-guided and genome mining strategies, ACS Sens, doi:10.1021/acssensors.1c02201
Toy, Gajic, Bacchetti, Looney, Gropper et al., Transfusion-related acute lung injury: incidence and risk factors, Blood, doi:10.1182/blood-2011-08-370932
Urban, Ermert, Schmid, Abu-Abed, Goosmann et al., Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans, PLoS Pathog, doi:10.1371/journal.ppat.1000639
Van Der Velden, Van Osch, Seghier, Bentlage, Mok et al., Complement C5a induces the generation of neutrophil extracellular traps by inhibiting mitochondrial STAT3 to promote the development of arterial thrombosis, Thromb J, doi:10.1186/s12959-022-00384-0
Van Hezel, Boshuizen, Peters, Straat, Vlaar et al., Red blood cell transfusion results in adhesion of neutrophils in human endotoxemia and in critically ill patients with sepsis, Transfusion, doi:10.1111/trf.15613
Van Raam, Sluiter, De Wit, Roos, Verhoeven et al., Mitochondrial membrane potential in human neutrophils is maintained by complex III activity in the absence of supercomplex organisation, PLoS One, doi:10.1371/journal.pone.0002013
Vlaar, Toy, Fung, Looney, Juffermans et al., A consensus redefinition of transfusion-related acute lung injury, Transfusion, doi:10.1111/trf.15311
Wang, Cao, Gorshkov, Zhou, Yang et al., Ablation of endothelial Pfkfb3 protects mice from acute lung injury in LPS-induced endotoxemia, Pharmacol Res, doi:10.1016/j.phrs.2019.104292
Wang, Li, Chen, Yuan, Su et al., GSDMD-dependent neutrophil extracellular traps promote macrophage-to-myofibroblast transition and renal fibrosis in obstructive nephropathy, Cell Death Dis, doi:10.1038/s41419-022-05138-4
Wang, Li, Stadler, Correll, Li et al., Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation, J Cell Biol, doi:10.1083/jcb.200806072
Wang, Wang, Zhao, Chen, Pluthero et al., NETosing neutrophils activate complement both on their own NETs and bacteria via alternative and non-alternative pathways, Clin Exp Immunol, doi:10.1159/000356980
Wang, Wu, Yan, Wang, Wu, M1-polarized alveolar macrophages are crucial in a mouse model of transfusion-related acute lung injury, Transfusion, doi:10.1111/trf.15609
Warnatsch, Ioannou, Wang, Papayannopoulos, Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis, Science, doi:10.1126/science.aaa8064
Wei, Zou, Xie, Wang, Huang et al., EDIL3 deficiency ameliorates adverse cardiac remodelling by neutrophil extracellular traps (NET)mediated macrophage polarization, Cardiovasc Res, doi:10.1093/cvr/cvab269
Wigerblad, Kaplan, Neutrophil extracellular traps in systemic autoimmune and autoinflammatory diseases, Nat Rev Immunol, doi:10.1038/s41577-022-00787-0
Wu, Huang, Hamanaka, Krause, Oh et al., HIF-1alpha is required for disturbed flow-induced metabolic reprogramming in human and porcine vascular endothelium, Elife, doi:10.7554/eLife.25217
Xie, Yang, Zhu, Gao, Jiang et al., Microparticles in red cell concentrates prime polymorphonuclear neutrophils and cause acute lung injury in a two-event mouse model, Int Immunopharmacol, doi:10.1016/j.intimp.2017.11.029
Xie, Zhu, Wan, Zhao, Meng et al., GSDMD-mediated NETosis promotes the development of acute respiratory distress syndrome, Eur J Immunol, doi:10.1002/eji.202250011
Xu, Zhang, Pelayo, Monestier, Ammollo et al., Extracellular histones are major mediators of death in sepsis, Nat Med, doi:10.1038/nm.2053
Yalcinkaya, Fotakis, Liu, Endo-Umeda, Dou et al., Cholesterol accumulation in macrophages drives NETosis in atherosclerotic plaques via IL-1beta secretion, Cardiovasc Res, doi:10.1093/cvr/cvac189
Yang, Duan, Liu, Xiong, Guan et al., A COX-2/ sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation, Theranostics, doi:10.7150/thno.43108
Yang, Feng, Chen, Wang, Chen et al., Disulfiram accelerates diabetic foot ulcer healing by blocking NET formation via suppressing the NLRP3/ Caspase-1/GSDMD pathway, Transl Res, doi:10.1016/j.trsl.2022.10.008
Yipp, Petri, Salina, Jenne, Scott et al., Infectioninduced NETosis is a dynamic process involving neutrophil multitasking in vivo, Nat Med, doi:10.1038/nm.2847
Yousefi, Mihalache, Kozlowski, Schmid, Hu, Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps, Cell Death Differ, doi:10.1038/cdd.2009.96
Zhang, Xu, Xu, Ye, Drug repurposing of ivermectin abrogates neutrophil extracellular traps and prevents melanoma metastasis, Front Oncol, doi:10.3389/fonc.2022.989167
Zhao, Liang, Ye, Wu, Qin et al., GSDMD promotes neutrophil extracellular traps via mtDNA-cGAS-STING pathway during lung ischemia/reperfusion, Cell Death Discov, doi:10.1038/s41420-023-01663-z
Zhao, Pan, Zhang, Ma, Zhang et al., Neutrophil extracellular traps in inflammatory bowel diseases: Implications in pathogenesis and therapeutic targets, Semin Cancer Biol, doi:10.1038/ncomms7673
Ziogas, Sajib, Lim, Alves, Das et al., PKM2 regulates endothelial cell junction dynamics and angiogenesis via ATP production, FASEB J, doi:10.1038/s41598-019-50866-x
Zuber, Fakhouri, Roumenina, Loirat, Vfrench et al., for a HCG. Use of eculizumab for atypical haemolytic uraemic syndrome and C3 glomerulopathies, Nat Rev Nephrol, doi:10.1038/nrneph.2012.214
Loading..
Please send us corrections, updates, or comments. c19early involves the extraction of 100,000+ datapoints from thousands of papers. Community updates help ensure high accuracy. Treatments and other interventions are complementary. All practical, effective, and safe means should be used based on risk/benefit analysis. No treatment or intervention is 100% available and effective for all current and future variants. We do not provide medical advice. Before taking any medication, consult a qualified physician who can provide personalized advice and details of risks and benefits based on your medical history and situation. FLCCC and WCH provide treatment protocols.
  or use drag and drop   
Submit