REPOZYTORIUM UNIWERSYTETU
W BIAŁYMSTOKU
UwB

Proszę używać tego identyfikatora do cytowań lub wstaw link do tej pozycji: http://hdl.handle.net/11320/16595
Pełny rekord metadanych
Pole DCWartośćJęzyk
dc.contributor.authorPiktel, Ewelina-
dc.contributor.authorWnorowska, Urszula-
dc.contributor.authorCieśluk, Mateusz-
dc.contributor.authorDeptuła, Piotr-
dc.contributor.authorPogoda, Katarzyna-
dc.contributor.authorMisztalewska-Turkowicz, Iwona-
dc.contributor.authorPaprocka, Paulina-
dc.contributor.authorNiemirowicz-Laskowska, Katarzyna-
dc.contributor.authorWilczewska, Agnieszka Z.-
dc.contributor.authorJanmey, Paul A.-
dc.contributor.authorBucki, Robert-
dc.date.accessioned2024-06-03T07:57:49Z-
dc.date.available2024-06-03T07:57:49Z-
dc.date.issued2019-
dc.identifier.citationJournal of Nanobiotechnology, Vol. 17 (2019), Article number: 22pl
dc.identifier.issn1477-3155-
dc.identifier.urihttp://hdl.handle.net/11320/16595-
dc.description.abstractBackground: Human plasma gelsolin (pGSN) is a multifunctional actin-binding protein involved in a variety of biological processes, including neutralization of pro-infammatory molecules such as lipopolysaccharide (LPS) and lipoteichoic acid (LTA) and modulation of host infammatory response. It was found that PBP10, a synthetic rhodamine B-conjugated peptide, based on the phosphoinositide-binding site of pGSN, exerts bactericidal activity against Grampositive and Gram-negative bacteria, interacts specifcally with LPS and LTA, and limits microbial-induced infammatory efects. The therapeutic efciency of PBP10 when immobilized on the surface of iron oxide-based magnetic nanoparticles was not evaluated, to date. Results: Using the human keratinocyte cell line HaCaT stimulated by bacterially-derived LPS and LTA as an in vitro model of bacterial infection, we examined the anti-infammatory efects of nanosystems consisting of iron oxidebased magnetic nanoparticles with aminosilane (MNP@NH2) or gold shells (MNP@Au) functionalized by a set of peptides, derived from the phosphatidylinositol 4,5-bisphosphate (PIP2)-binding site of the human plasma protein gelsolin, which also binds LPS and LTA. Our results indicate that these nanosystems can kill both Gram-positive and Gram-negative bacteria and limit the production of infammatory mediators, including nitric oxide (NO), reactive oxygen species (ROS), and interleukin-8 (IL-8) in the response to heat-killed microbes or extracted bacterial cell wall components. The nanoparticles possess the potential to improve therapeutic efcacy and are characterized by lower toxicity and improved hemocompatibility when compared to free peptides. Atomic force microscopy (AFM) showed that these PBP10-based nanosystems prevented changes in nanomechanical properties of cells that were otherwise stimulated by LPS. Conclusions: Neutralization of endotoxemia-mediated cellular efects by gelsolin-derived peptides and PBP10-containing nanosystems might be considered as potent therapeutic agents in the improved therapy of bacterial infections and microbial-induced infammation.pl
dc.description.sponsorshipThis work was fnancially supported by the National Science Center, Poland under Grant: UMO-2015/17/B/NZ6/03473 (to RB) and Medical University of Bialystok (N/ST/ZB/18/002/1162 and N/ST/ZB/18/001/1162 (to RB) and N/ST/MN/18/002/1162 (to EP). Part of the study was conducted with the use of equipment purchased by the Medical University of Białystok as part of the RPOWP 2007-2013 funding, Priority I, Axis 1.1, contract No. UDARPPD.01.01.00-20-001/15-00 dated 26.06.2015. The physicochemical studies were performed in Centre of Synthesis and Analysis BioNanoTechno of the University of Bialystok (POPW.01.03.00-20-034/09-00 and POPW.01.03.00-20004/11 projects). EP acknowledges a doctoral scholarship from Polpharma Scientifc Foundation, Poland. PAJ and RB acknowledge support from NIH grant GM111942-01.pl
dc.language.isoenpl
dc.publisherBioMed Central (BMC)pl
dc.rightsUznanie autorstwa 4.0 Międzynarodowe*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/*
dc.subjectGelsolinpl
dc.subjectInfammationpl
dc.subjectSkin diseasespl
dc.subjectPBP10pl
dc.subjectMagnetic nanoparticlespl
dc.titleInhibition of inflammatory response in human keratinocytes by magnetic nanoparticles functionalized with PBP10 peptide derived from the PIP2-binding site of human plasma gelsolinpl
dc.typeArticlepl
dc.rights.holder© The Author(s) 2019. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/)pl
dc.identifier.doi10.1186/s12951-019-0455-5-
dc.description.EmailRobert Bucki: buckirobert@gmail.compl
dc.description.AffiliationEwelina Piktel - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationUrszula Wnorowska - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationMateusz Cieśluk - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationPiotr Deptula - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationKatarzyna Pogoda - IInstitute of Nuclear Physics Polish Academy of Sciencespl
dc.description.AffiliationIwona Misztalewska‑Turkowicz - Institute of Chemistry, University of Białystokpl
dc.description.AffiliationPaulina Paprocka - Department of Microbiology and Immunology, The Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielcepl
dc.description.AffiliationKatarzyna Niemirowicz‑Laskowska - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationAgnieszka Z. Wilczewska - Institute of Chemistry, University of Białystokpl
dc.description.AffiliationPaul A. Janmey - Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvaniapl
dc.description.AffiliationRobert Bucki - Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.referencesSalmon JK, Armstrong CA, Ansel JC. The skin as an immune organ. West J Med. 1994;160(2):146–52pl
dc.description.referencesBarker JN, Mitra RS, Grifths CE, Dixit VM, Nickolof BJ. Keratinocytes as initiators of infammation. Lancet. 1991;337(8735):211–4.pl
dc.description.referencesKarin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial infections to chronic infammation and cancer. Cell. 2006;124(4):823–35.pl
dc.description.referencesKim MY, Lim YY, Kim HM, Park YM, Kang H, Kim BJ. Synergistic inhibition of tumor necrosis factor-alpha-stimulated pro-infammatory cytokine expression in HaCaT cells by a combination of rapamycin and mycophenolic acid. Ann Dermatol. 2015;27(1):32–9.pl
dc.description.referencesGutsmann T, Razquin-Olazarán I, Kowalski I, Kaconis Y, Howe J, Bartels R, et al. New antiseptic peptides to protect against endotoxin-mediated shock. Antimicrob Agents Chemother. 2010;54(9):3817–24.pl
dc.description.referencesPfalzgraf A, Heinbockel L, Su Q, Gutsmann T, Brandenburg K, Weindl G. Synthetic antimicrobial and LPS-neutralising peptides suppress infammatory and immune responses in skin cells and promote keratinocyte migration. Sci Rep. 2016;6:31577.pl
dc.description.referencesEckmann C, Dryden M. Treatment of complicated skin and soft-tissue infections caused by resistant bacteria: value of linezolid, tigecycline, daptomycin and vancomycin. Eur J Med Res. 2010;15(12):554–63.pl
dc.description.referencesBucki R, Byfeld FJ, Kulakowska A, McCormick ME, Drozdowski W, Namiot Z, et al. Extracellular gelsolin binds lipoteichoic acid and modulates cellular response to proinfammatory bacterial wall components. J Immunol. 2008;181(7):4936–44pl
dc.description.referencesBucki R, Georges PC, Espinassous Q, Funaki M, Pastore JJ, Chaby R, et al. Inactivation of endotoxin by human plasma gelsolin. Biochemistry. 2005;44(28):9590–7.pl
dc.description.referencesBucki R, Kulakowska A, Byfeld FJ, Zendzian-Piotrowska M, Baranowski M, Marzec M, et al. Plasma gelsolin modulates cellular response to sphingosine 1-phosphate. Am J Physiol Cell Physiol. 2010;299(6):C1516–23.pl
dc.description.referencesWątek M, Durnaś B, Wollny T, Pasiarski M, Góźdź S, Marzec M, et al. Unexpected profle of sphingolipid contents in blood and bone marrow plasma collected from patients diagnosed with acute myeloid leukemia. Lipids Health Dis. 2017;16(1):235.pl
dc.description.referencesBucki R, Levental I, Kulakowska A, Janmey PA. Plasma gelsolin: function, prognostic value, and potential therapeutic use. Curr Protein Pept Sci. 2008;9(6):541–51.pl
dc.description.referencesPiktel E, Levental I, Durnas B, Janmey PA, Bucki R. Plasma gelsolin: indicator of infammation and its potential as a diagnostic tool and therapeutic target. Int J Mol Sci. 2018;19(9):2516.pl
dc.description.referencesBucki R, Janmey PA. Interaction of the gelsolin-derived antibacterial PBP 10 peptide with lipid bilayers and cell membranes. Antimicrob Agents Chemother. 2006;50(9):2932–40.pl
dc.description.referencesFu H, Björkman L, Janmey P, Karlsson A, Karlsson J, Movitz C, et al. The two neutrophil members of the formylpeptide receptor family activate the NADPH-oxidase through signals that difer in sensitivity to a gelsolin derived phosphoinositide-binding peptide. BMC Cell Biol. 2004;5(1):50.pl
dc.description.referencesLi Y, Liu W, Sun C, Zheng M, Zhang J, Liu B, et al. Hybrids of carbon dots with subunit B of ricin toxin for enhanced immunomodulatory activity. J Colloid Interface Sci. 2018;523:226–33.pl
dc.description.referencesNikapitiya C, Dananjaya SHS, De Silva BCJ, Heo GJ, Oh C, De Zoysa M, et al. Chitosan nanoparticles: a positive immune response modulator as display in zebrafsh larvae against Aeromonas hydrophila infection. Fish Shellfsh Immunol. 2018;76:240–6.pl
dc.description.referencesLappas CM. The immunomodulatory efects of titanium dioxide and silver nanoparticles. Food Chem Toxicol. 2015;85:78–83.pl
dc.description.referencesMa JS, Kim WJ, Kim JJ, Kim TJ, Ye SK, Song MD, et al. Gold nanoparticles attenuate LPS-induced NO production through the inhibition of NFkappaB and IFN-beta/STAT1 pathways in RAW264.7 cells. Nitric Oxide. 2010;23(3):214–9.pl
dc.description.referencesGatto F, Moglianetti M, Pompa PP, Bardi G. Platinum nanoparticles decrease reactive oxygen species and modulate gene expression without alteration of immune responses in THP-1 monocytes. Nanomaterials. 2018;8(6):392.pl
dc.description.referencesPrasad P, Sachan S, Suman S, Swayambhu G, Gupta S. Regenerative core-shell nanoparticles for simultaneous removal and detection of endotoxins. Langmuir. 2018;34(25):7396–403.pl
dc.description.referencesMoyano DF, Liu Y, Ayaz F, Hou S, Puangploy P, Duncan B, et al. Immunomodulatory efects of coated gold nanoparticles in LPS-stimulated. Chem. 2016;1(2):320–7pl
dc.description.referencesPereira DV, Petronilho F, Pereira HR, Vuolo F, Mina F, Possato JC, et al. Efects of gold nanoparticles on endotoxin-induced uveitis in rats. Invest Ophthalmol Vis Sci. 2012;53(13):8036–41.pl
dc.description.referencesGregory AE, Judy BM, Qazi O, Blumentritt CA, Brown KA, Shaw AM, et al. A gold nanoparticle-linked glycoconjugate vaccine against Burkholderia mallei. Nanomedicine. 2015;11(2):447–56.pl
dc.description.referencesNiemirowicz-Laskowska K, Głuszek K, Piktel E, Pajuste K, Durnaś B, Król G, et al. Bactericidal and immunomodulatory properties of magnetic nanoparticles functionalized by 1,4-dihydropyridines. Int J Nanomed. 2018;13:3411–24.pl
dc.description.referencesMassart R. Preparation of aqueous magnetic liquids in alkaline and acidic media. IEEE Trans Magn. 1981;17(2):1247–8.pl
dc.description.referencesNiemirowicz K, Swiecicka I, Wilczewska AZ, Misztalewska I, Kalska-Szostko B, Bienias K, et al. Gold-functionalized magnetic nanoparticles restrict growth of Pseudomonas aeruginosa. Int J Nanomed. 2014;9:2217–24.pl
dc.description.referencesDehghani H, Hashemi M, Entezari M, Mohsenifar A. The comparison of anticancer activity of thymoquinone and nanothymoquinone on human breast adenocarcinoma. Iran J Pharm Res. 2015;14(2):539–46.pl
dc.description.referencesDong N, Ma Q, Shan A, Lv Y, Hu W, Gu Y, et al. Strand length-dependent antimicrobial activity and membrane-active mechanism of arginine- and valine-rich β-hairpin-like antimicrobial peptides. Antimicrob Agents Chemother. 2012;56(6):2994–3003.pl
dc.description.referencesDurnaś B, Wnorowska U, Pogoda K, Deptuła P, Wątek M, Piktel E, et al. Candidacidal activity of selected ceragenins and human cathelicidin LL-37 in experimental settings mimicking infection sites. PLoS ONE. 2016;11(6):e0157242pl
dc.description.referencesYoon WJ, Kim SS, Oh TH, Lee NH, Hyun CG. Abies koreana essential oil inhibits drug-resistant skin pathogen growth and LPS-induced infammatory efects of murine macrophage. Lipids. 2009;44(5):471–6.pl
dc.description.referencesSchwaminger SP, Garcia PF, Merck GK, Bodensteiner FA, Heissler S, Sebastian G, et al. Nature of interactions of amino acids with bare magnetite nanoparticles. J Phys Chem. 2015;119(40):23032–41.pl
dc.description.referencesNiemirowicz K, Surel U, Wilczewska AZ, Mystkowska J, Piktel E, Gu X, et al. Bactericidal activity and biocompatibility of ceragenin-coated magnetic nanoparticles. J Nanobiotechnol. 2015;13:32.pl
dc.description.referencesBucki R, Pastore JJ, Randhawa P, Vegners R, Weiner DJ, Janmey PA. Antibacterial activities of rhodamine B-conjugated gelsolin-derived peptides compared to those of the antimicrobial peptides catheli‑cidin LL37, magainin II, and melittin. Antimicrob Agents Chemother. 2004;48(5):1526–33.pl
dc.description.referencesLaskin JD, Heck DE, Laskin DL. Multifunctional role of nitric oxide in infammation. Trends Endocrinol Metab. 1994;5(9):377–82.pl
dc.description.referencesYoung CN, Koepke JI, Terlecky LJ, Borkin MS, Boyd Savoy L, Boyd SL, et al. Reactive oxygen species in tumor necrosis factor-alpha-activated primary human keratinocytes: implications for psoriasis and infammatory skin disease. J Invest Dermatol. 2008;128(11):2606–14.pl
dc.description.referencesSticherling M, Bornscheuer E, Schröder JM, Christophers E. Localization of neutrophil-activating peptide-1/interleukin-8-immunoreactivity in normal and psoriatic skin. J Invest Dermatol. 1991;96(1):26–30.pl
dc.description.referencesPi J, Li T, Liu J, Su X, Wang R, Yang F, et al. Detection of lipopolysaccharide induced infammatory responses in RAW264.7 macrophages using atomic force microscope. Micron. 2014;65:1–9.pl
dc.description.referencesPi J, Cai H, Yang F, Jin H, Liu J, Yang P, et al. Atomic force microscopy based investigations of anti-infammatory efects in lipopolysaccharide-stimulated macrophages. Anal Bioanal Chem. 2016;408(1):165–76.pl
dc.description.referencesKaminska PS, Yernazarova A, Murawska E, Swiecicki J, Fiedoruk K, Bideshi DK, et al. Comparative analysis of quantitative reverse transcription real-time PCR and commercial enzyme imunoassays for detection of enterotoxigenic Bacillus thuringiensis isolates. FEMS Microbiol Lett. 2014;357(1):34–9.pl
dc.description.referencesGustafsson A, Olin AI, Ljunggren L. LPS interactions with immobilized and soluble antimicrobial peptides. Scand J Clin Lab Invest. 2010;70(3):194–200.pl
dc.description.referencesScott A, Weldon S, Buchanan PJ, Schock B, Ernst RK, McAuley DF, et al. Evaluation of the ability of LL-37 to neutralise LPS in vitro and ex vivo. PLoS ONE. 2011;6(10):e26525.pl
dc.description.referencesWitke W, Sharpe AH, Hartwig JH, Azuma T, Stossel TP, Kwiatkowski DJ. Hemostatic, infammatory, and fbroblast responses are blunted in mice lacking gelsolin. Cell. 1995;81(1):41–51.pl
dc.description.referencesChristofdou-Solomidou M, Scherpereel A, Solomides CC, Muzykantov VR, Machtay M, Albelda SM, et al. Changes in plasma gelsolin concentration during acute oxidant lung injury in mice. Lung. 2002;180(2):91–104.pl
dc.description.referencesYang Z, Chiou TT, Stossel TP, Kobzik L. Plasma gelsolin improves lung host defense against pneumonia by enhancing macrophage NOS3 function. Am J Physiol Lung Cell Mol Physiol. 2015;309(1):L11–6.pl
dc.description.referencesOrdija CM, Chiou TT, Yang Z, Deloid GM, de Oliveira Valdo M, Wang Z, et al. Free actin impairs macrophage bacterial defenses via scavenger receptor MARCO interaction with reversal by plasma gelsolin. Am J Physiol Lung Cell Mol Physiol. 2017;312(6):L1018–28.pl
dc.description.referencesTaylor E, Webster TJ. Reducing infections through nanotechnology and nanoparticles. Int J Nanomedicine. 2011;6:1463–73.pl
dc.description.referencesLiu Z, Li W, Wang F, Sun C, Wang L, Wang J, et al. Enhancement of lipopolysaccharide-induced nitric oxide and interleukin-6 production by PEGylated gold nanoparticles in RAW264.7 cells. Nanoscale. 2012;4(22):7135–42.pl
dc.description.referencesNishanth RP, Jyotsna RG, Schlager JJ, Hussain SM, Reddanna P. Infammatory responses of RAW 264.7 macrophages upon exposure to nanoparticles: role of ROS-NFκB signaling pathway. Nanotoxicology. 2011;5(4):502–16.pl
dc.description.referencesNiemirowicz K, Surel U, Wilczewska AZ, Mystkowska J, Piktel E, Gu X, et al. Bactericidal activity and biocompatibility of ceragenin-coated magnetic nanoparticles. J Nanobiotechnol. 2015;13(1):32.pl
dc.description.referencesLee WR, Kim KH, An HJ, Kim JY, Chang YC, Chung H, et al. The protective efects of melittin on Propionibacterium acnes-induced infammatory responses in vitro and in vivo. J Invest Dermatol. 2014;134(7):1922–30pl
dc.description.referencesMohamed MF, Seleem MN. Efcacy of short novel antimicrobial and anti-infammatory peptides in a mouse model of methicillin-resistant Staphylococcus aureus (MRSA) skin infection. Drug Des Devel Ther. 2014;8:1979–83.pl
dc.description.referencesCarretero M, Escámez MJ, García M, Duarte B, Holguín A, Retamosa L, et al. In vitro and in vivo wound healing-promoting activities of human cathelicidin LL-37. J Invest Dermatol. 2008;128(1):223–36.pl
dc.description.referencesFiedoruk K, Daniluk T, Rozkiewicz D, Zaremba ML, Oldak E, Sciepuk M, et al. Conventional and molecular methods in the diagnosis of community-acquired diarrhoea in children under 5 years of age from the north-eastern region of Poland. Int J Infect Dis. 2015;37:145–51.pl
dc.description.referencesMatsuzaki K, Sugishita K, Miyajima K. Interactions of an antimicrobial peptide, magainin 2, with lipopolysaccharide-containing liposomes as a model for outer membranes of Gram-negative bacteria. FEBS Lett. 1999;449(2–3):221–4.pl
dc.description.referencesScott MG, Gold MR, Hancock RE. Interaction of cationic peptides with lipoteichoic acid and Gram-positive bacteria. Infect Immun. 1999;67(12):6445–53.pl
dc.description.referencesWang B, McHugh BJ, Qureshi A, Campopiano DJ, Clarke DJ, Fitzgerald JR, et al. IL-1β-induced protection of keratinocytes against Staphylococcus aureus-secreted proteases is mediated by human β-defensin 2. J Invest Dermatol. 2017;137(1):95–105.pl
dc.description.referencesMarcatili A, de Cipollaro l’Ero G, Galdiero M, Folgore A, Petrillo G. TNFalpha, IL-1 alpha, IL-6 and ICAM-1 expression in human keratinocytes stimulated in vitro with Escherichia coli heat-shock proteins. Microbiology. 1997;143:45–53.pl
dc.description.referencesLai Y, Gallo RL. Toll-like receptors in skin infections and infammatory diseases. Infect Disord Drug Targets. 2008;8(3):144–55.pl
dc.description.referencesArany I, Brysk MM, Brysk H, Tyring SK. Regulation of inducible nitric oxide synthase mRNA levels by diferentiation and cytokines in human keratinocytes. Biochem Biophys Res Commun. 1996;220(3):618–22.pl
dc.description.referencesGoldsmith PC, Leslie TA, Hayes NA, Levell NJ, Dowd PM, Foreman JC. Inhibitors of nitric oxide synthase in human skin. J Invest Dermatol. 1996;106(1):113–8.pl
dc.description.referencesHerman AG, Moncada S. Therapeutic potential of nitric oxide donors in the prevention and treatment of atherosclerosis. Eur Heart J. 2005;26(19):1945–55.pl
dc.description.referencesWong VW, Lerner E. Nitric oxide inhibition strategies. Future Sci OA. 2015. https://doi.org/10.4155/fso.15.35.pl
dc.description.referencesBécherel PA, Le Gof L, Ktorza S, Chosidow O, Francès C, Issaly F, et al. CD23-mediated nitric oxide synthase pathway induction in human keratinocytes is inhibited by retinoic acid derivatives. J Invest Dermatol. 1996;106(6):1182–6.pl
dc.description.referencesNakashima T, Sato E, Niwano Y, Kohno M, Muraoka W, Oda T. Inhibitory or scavenging action of ketoconazole and ciclopiroxolamine against reactive oxygen species released by primed infammatory cells. Br J Dermatol. 2007;156(4):720–7.pl
dc.description.referencesGrange PA, Chéreau C, Raingeaud J, Nicco C, Weill B, Dupin N, et al. Production of superoxide anions by keratinocytes initiates P. acnes-induced infammation of the skin. PLoS Pathog. 2009;5(7):1000527.pl
dc.description.referencesDing W, Hudson LG, Liu KJ. Inorganic arsenic compounds cause oxidative damage to DNA and protein by inducing ROS and RNS generation in human keratinocytes. Mol Cell Biochem. 2005;279(1–2):105–12.pl
dc.description.referencesBruch-Gerharz D, Fehsel K, Suschek C, Michel G, Ruzicka T, Kolb-Bachofen V. A proinfammatory activity of interleukin 8 in human skin: expression of the inducible nitric oxide synthase in psoriatic lesions and cultured keratinocytes. J Exp Med. 1996;184(5):2007–12.pl
dc.description.referencesPogoda K, Jaczewska J, Wiltowska-Zuber J, Klymenko O, Zuber K, Fornal M, et al. Depth-sensing analysis of cytoskeleton organization based on AFM data. Eur Biophys J. 2012;41(1):79–87.pl
dc.description.referencesLeporatti S, Gerth A, Köhler G, Kohlstrunk B, Hauschildt S, Donath E. Elasticity and adhesion of resting and lipopolysaccharide-stimulated macrophages. FEBS Lett. 2006;580(2):450–4.pl
dc.description.referencesRoca-Cusachs P, Almendros I, Sunyer R, Gavara N, Farré R, Navajas D. Rheology of passive and adhesion-activated neutrophils probed by atomic force microscopy. Biophys J. 2006;91(9):3508–18.pl
dc.description.referencesMeng F, Mambetsariev I, Tian Y, Beckham Y, Meliton A, Lef A, et al. Attenuation of lipopolysaccharide-induced lung vascular stifening by lipoxin reduces lung infammation. Am J Respir Cell Mol Biol. 2015;52(2):152–61.pl
dc.description.referencesByfeld FJ, Kowalski M, Cruz K, Leszczynska K, Namiot A, Savage PB, et al. Cathelicidin LL-37 increases lung epithelial cell stifness, decreases transepithelial permeability, and prevents epithelial invasion by Pseudomonas aeruginosa. J Immunol. 2011;187(12):6402–9.pl
dc.description.volume17pl
dc.identifier.citation2Journal of Nanobiotechnologypl
Występuje w kolekcji(ach):Artykuły naukowe (WChem)

Pokaż uproszczony widok rekordu Zobacz statystyki


Pozycja ta dostępna jest na podstawie licencji Licencja Creative Commons CCL Creative Commons