Vis enkel innførsel

dc.contributor.authorGerogianni, Alexandra
dc.contributor.authorBal, Melissa
dc.contributor.authorMohlin, Camilla
dc.contributor.authorWoodruff, Trent M.
dc.contributor.authorLambris, John D.
dc.contributor.authorMollnes, Tom Eirik
dc.contributor.authorSjöström, Dick J.
dc.contributor.authorNilsson, Per
dc.date.accessioned2023-10-31T08:30:48Z
dc.date.available2023-10-31T08:30:48Z
dc.date.created2023-05-22T13:53:24Z
dc.date.issued2023
dc.identifier.citationFrontiers in Immunology. 2023, 14 .en_US
dc.identifier.issn1664-3224
dc.identifier.urihttps://hdl.handle.net/11250/3099625
dc.description.abstractIron oxide nanoparticles (IONPs) are widely used in diagnostic and therapeutic settings. Upon systemic administration, however, they are rapidly recognized by components of innate immunity, which limit their therapeutic capacity and can potentially lead to adverse side effects. IONPs were previously found to induce the inflammatory response in human whole blood, including activation of the complement system and increased secretion of cytokines. Here, we investigated the thromboinflammatory response of 10-30 nm IONPs in lepirudin anticoagulated whole blood in interplay with endothelial cells and evaluated the therapeutic effect of applying complement inhibitors to limit adverse effects related to thromboinflammation. We found that IONPs induced complement activation, primarily at the C3-level, in whole blood incubated for up to four hours at 37°C with and without human microvascular endothelial cells. Furthermore, IONPs mediated a strong thromboinflammatory response, as seen by the significantly increased release of 21 of the 27 analyzed cytokines (p<0.05). IONPs also significantly increased cell-activation markers of endothelial cells [ICAM-1 (p<0.0001), P/E-selectin (p<0.05)], monocytes, and granulocytes [CD11b (p<0.001)], and platelets [CD62P (p<0.05), CD63 (p<0.05), NAP-2 (p<0.01), PF4 (p<0.05)], and showed cytotoxic effects, as seen by increased LDH (p<0.001) and heme (p<0.0001) levels. We found that inflammation and endothelial cell activation were partly complement-dependent and inhibition of complement at the level of C3 by compstatin Cp40 significantly attenuated expression of ICAM-1 (p<0.01) and selectins (p<0.05). We show that complement activation plays an important role in the IONPs-induced thromboinflammatory response and that complement inhibition is promising in improving IONPs biocompatibility.en_US
dc.language.isoengen_US
dc.publisherFrontiers Media S. A.en_US
dc.rightsNavngivelse 4.0 Internasjonal*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/deed.no*
dc.titleIn vitro evaluation of iron oxide nanoparticle-induced thromboinflammatory response using a combined human whole blood and endothelial cell modelen_US
dc.title.alternativeIn vitro evaluation of iron oxide nanoparticle-induced thromboinflammatory response using a combined human whole blood and endothelial cell modelen_US
dc.typePeer revieweden_US
dc.typeJournal articleen_US
dc.description.versionpublishedVersionen_US
dc.source.volume14en_US
dc.source.journalFrontiers in Immunologyen_US
dc.identifier.doi10.3389/fimmu.2023.1101387
dc.identifier.cristin2148515
cristin.ispublishedtrue
cristin.fulltextoriginal
cristin.qualitycode1


Tilhørende fil(er)

Thumbnail

Denne innførselen finnes i følgende samling(er)

Vis enkel innførsel

Navngivelse 4.0 Internasjonal
Med mindre annet er angitt, så er denne innførselen lisensiert som Navngivelse 4.0 Internasjonal