REPOZYTORIUM UNIWERSYTETU
W BIAŁYMSTOKU
UwB

Proszę używać tego identyfikatora do cytowań lub wstaw link do tej pozycji: http://hdl.handle.net/11320/16573
Pełny rekord metadanych
Pole DCWartośćJęzyk
dc.contributor.authorPiktel, Ewelina-
dc.contributor.authorMarkiewicz, Karolina H.-
dc.contributor.authorWilczewska, Agnieszka Z.-
dc.contributor.authorDaniluk, Tamara-
dc.contributor.authorChmielewska, Sylwia-
dc.contributor.authorNiemirowicz-Laskowska, Katarzyna-
dc.contributor.authorMystkowska, Joanna-
dc.contributor.authorPaprocka, Paulina-
dc.contributor.authorSavage, Paul B.-
dc.contributor.authorBucki, Robert-
dc.date.accessioned2024-05-28T10:34:50Z-
dc.date.available2024-05-28T10:34:50Z-
dc.date.issued2020-
dc.identifier.citationInternational Journal of Nanomedicine, Volume 15 (2020), p. 4573-4589pl
dc.identifier.issn1176-9114 (Print)-
dc.identifier.urihttp://hdl.handle.net/11320/16573-
dc.description.abstractBackground: Therapeutic efficiency of ceragenins against cancers may be limited by lack of their hemocompatibility when high concentrations of molecules are required to reach a desired result. Synergistic effects observed upon administration of anticancer agents and metal nanoparticles may provide an opportunity to limit toxicity of immobilized ceragenins on the surface of metal nanoparticles and to improve their therapeutic efficiency at the same time. The aim of present work is to investigate the anticancer activities and hemocompatibility of nanoformulations consisting of ceragenin CSA-131 united with aminosilanemodified iron oxide-based magnetic nanoparticles (MNP) and prepared by 1) covalent bonding (MNP@CSA-131) or 2) by combining CSA-131 with MNP in 1:1 ratio (CSA-131 + MNP). Possible synergistic interactions between CSA-131 and magnetic nanoparticles were also quantified.pl
dc.description.abstractMethods: MNP@CSA-131 and CSA-131+MNP were tested in vitro against selected lung and colon cancer cells using colorimetric, fluorimetric and flow cytometry methods.pl
dc.description.abstractResults: Performed analysis demonstrates that MNP-based nanosystems significantly improve the killing efficiency of tested ceragenin, decreasing the viability of extra 1.37±4.72% to 76.07±15.30% cancer cells when compared to free CSA-131. Quantification of synergistic effects indicates the favorable interactions between CSA-131 and magnetic nanoparticles (CI < 1 for all tested doses), revealing at the same time a reduction in effective doses of ceragenin from 1.17 ±0.61 to 34.57 ± 12.78 times when combined with MNP. We demonstrate that both MNP@CSA131 and CSA-131+MNP induce significantly apoptosis of cancer cells and prevent the division of colon cancer cells even at relatively low doses of the active compound (10 µg/mL). Importantly, combining CSA-131 with MNP decreases the hemolytic activity of free ceragenin 4.72 to 7.88 times, which indicates a considerable improvement of hemotoxicity profile.pl
dc.description.abstractConclusion: Comparative analyses have revealed that both developed CSA-containing nanoformulations due to the utility of synergistic interactions between MNP and CSA-131, which are effective against lung and colon cancer cells. This indicates the new directions in preparation of MNP-based therapeutics, which are relatively easy to synthetize, costeffective and safe when intravenously administrated.pl
dc.description.sponsorshipThis work was financially supported by grants from the National Science Centre, Poland (UMO-2015/19/N/NZ6/01872 to EP) and Medical University of Bialystok (SUB/1/DN/19/001/1162 to RB). 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. UDA- RPPD.01.01.00-20-001/15-00 dated 26.06.2015. The synthesis and physicochemical analysis of magnetic nanoparticles and MNP-based compounds were performed in the Centre of Synthesis and Analysis BioNanoTechno of the University of Bialystok (POPW.01.03.00-20-034/09-00 and POPW.01.03.00-20-004/11 projects). This work was supported by the program of the Minister of Science and Higher Education under the name “Regional Initiative of Excellence in 2019-2022", project number: 024/RID/2018/19, financing amount: 11.999.000,00 PLN. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.pl
dc.language.isoenpl
dc.publisherDove Medical Presspl
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 Międzynarodowe*
dc.rightsUznanie autorstwa-Użycie niekomercyjne 4.0 Międzynarodowe*
dc.rights.urihttp://creativecommons.org/licenses/by-nc/4.0/*
dc.subjectcerageninspl
dc.subjectanticancer activitypl
dc.subjectcolon cancerpl
dc.subjectlung cancerpl
dc.subjectsynergistic effectspl
dc.subjectcombinatory therapypl
dc.titleQuantification of Synergistic Effects of Ceragenin CSA-131 Combined with Iron Oxide Magnetic Nanoparticles Against Cancer Cellspl
dc.typeArticlepl
dc.rights.holder© 2020 Piktel et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms. php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/).pl
dc.identifier.doi10.2147/IJN.S255170-
dc.description.EmailRobert Bucki: buckirobert@gmail.compl
dc.description.AffiliationEwelina Piktel - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationKarolina H. Markiewicz - Faculty of Chemistry, University of Bialystokpl
dc.description.AffiliationAgnieszka Z. Wilczewska - Faculty of Chemistry, University of Bialystokpl
dc.description.AffiliationTamara Daniluk - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationSylwia Chmielewska - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationKatarzyna Niemirowicz-Laskowska - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystokpl
dc.description.AffiliationJoanna Mystkowska - Department of Materials and Biomedical Engineering, Białystok University of Technologypl
dc.description.AffiliationPaulina Paprocka - Department of Microbiology and Immunology, The Faculty of Medicine and Health Sciences, Jan Kochanowski University in Kielcepl
dc.description.AffiliationPaul B. Savage - Department of Chemistry and Biochemistry, Brigham Young Universitypl
dc.description.AffiliationRobert Bucki - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok; Department of Microbiology and Immunology, The Faculty of Medicine and Health Sciences, Jan Kochanowski University in Kielcepl
dc.description.referencesFelício MR, Silva ON, Gonçalves S, Santos NC, Franco OL. Peptides with dual antimicrobial and anticancer activities. Front Chem. 2017;5:5. doi:10.3389/fchem.2017.00005pl
dc.description.referencesDeslouches B, Di YP. Antimicrobial peptides with selective antitumor mechanisms: prospect for anticancer applications. Oncotarget. 2017;8(28):46635–46651. doi:10.18632/oncotarget.16743pl
dc.description.referencesBaxter AA, Lay FT, Poon IKH, Kvansakul M, Hulett MD. Tumor cell membrane-targeting cationic antimicrobial peptides: novel insights into mechanisms of action and therapeutic prospects. Cell Mol Life Sci. 2017;74(20):3809–3825. doi:10.1007/s00018-017-2604-zpl
dc.description.referencesPiktel E, Prokop I, Wnorowska U, et al. Ceragenin CSA-13 as free molecules and attached to magnetic nanoparticle surfaces induce caspase-dependent apoptosis in human breast cancer cells via disruption of cell oxidative balance. Oncotarget. 2018;9(31):21904–21920. doi:10.18632/oncotarget.25105pl
dc.description.referencesKuroda K, Fukuda T, Okumura K, et al. Ceragenin CSA-13 induces cell cycle arrest and antiproliferative effects in wild-type and p53 null mutant HCT116 colon cancer cells. Anticancer Drugs. 2013;24 (8):826–834. doi:10.1097/CAD.0b013e3283634dd0pl
dc.description.referencesNiemirowicz K, Prokop I, Wilczewska AZ, et al. Magnetic nanoparticles enhance the anticancer activity of cathelicidin LL-37 peptide against colon cancer cells. Int J Nanomedicine. 2015;10:3843–3853. doi:10.2147/IJN.S76104pl
dc.description.referencesNiemirowicz K, Surel U, Wilczewska AZ, et al. Bactericidal activity and biocompatibility of ceragenin-coated magnetic nanoparticles. J Nanobiotechnol. 2015;13(1):32. doi:10.1186/s12951-015-0093-5pl
dc.description.referencesNavya PN, Kaphle A, Srinivas SP, Bhargava SK, Rotello VM, Daima HK. Current trends and challenges in cancer management and therapy using designer nanomaterials. Nano Converg. 2019;6 (1):23.pl
dc.description.referencesNiemirowicz K, Markiewicz KH, Wilczewska AZ, Car H. Magnetic nanoparticles as new diagnostic tools in medicine. Adv Med Sci. 2012;57(2):196–207. doi:10.2478/v10039-012-0031-9pl
dc.description.referencesTokajuk G, Niemirowicz K, Deptula P, et al. Use of magnetic nanoparticles as a drug delivery system to improve chlorhexidine antimicrobial activity. Int J Nanomedicine. 2017;12:7833–7846. doi:10.2147/IJN.S140661pl
dc.description.referencesFathima JB, Pugazhendhi A, Venis R. Synthesis and characterization of ZrO. Microb Pathog. 2017;110:245–251. doi:10.1016/j.micpath.2017.06.039pl
dc.description.referencesSathiyavimal S, Vasantharaj S, Bharathi D, et al. Biogenesis of copper oxide nanoparticles (CuONPs) using Sida acuta and their incorporation over cotton fabrics to prevent the pathogenicity of Gram negative and Gram positive bacteria. J Photochem Photobiol B. 2018;188:126–134. doi:10.1016/j.jphotobiol.2018.09.014pl
dc.description.referencesPugazhendhi A, Prabhu R, Muruganantham K, Shanmuganathan R, Natarajan S. Anticancer, antimicrobial and photocatalytic activities of green synthesized magnesium oxide nanoparticles (MgONPs) using aqueous extract of Sargassum wightii. J Photochem Photobiol B. 2019;190:86–97. doi:10.1016/j.jphotobiol.2018.11.014pl
dc.description.referencesChen Z, Zheng Y, Shi Y, Cui Z. Overcoming tumor cell chemoresistance using nanoparticles: lysosomes are beneficial for (stearoyl) gemcitabine-incorporated solid lipid nanoparticles. Int J Nanomedicine. 2018;13:319–336. doi:10.2147/IJN.S149196pl
dc.description.referencesKouassi GK, Irudayaraj J. Magnetic and gold-coated magnetic nanoparticles as a DNA sensor. Anal Chem. 2006;78(10):3234–3241. doi:10.1021/ac051621jpl
dc.description.referencesMaddinedi SB. Green synthesis of Au–Cu2−xSe heterodimer nanoparticles and their in-vitro cytotoxicity, photothermal assay. Environ Toxicol Pharmacol. 2017;53:29–33. doi:10.1016/j.etap.2017.05.006pl
dc.description.referencesMaddinedi SB, Mandal BK, Anna KK. Tyrosine assisted size controlled synthesis of silver nanoparticles and their catalytic, in-vitro cytotoxicity evaluation. Environ Toxicol Pharmacol. 2017;51:23–29. doi:10.1016/j.etap.2017.02.020pl
dc.description.referencesMaddinedi SB, Mandal BK, Maddili SK. Biofabrication of size controllable silver nanoparticles - A green approach. J Photochem Photobiol B. 2017;167:236–241. doi:10.1016/j.jphotobiol.2017.01.003pl
dc.description.referencesMaddinedi SB, Mandal BK, Anna KK. Environment friendly approach for size controllable synthesis of biocompatible Silver nanoparticles using diastase. Environ Toxicol Pharmacol. 2017;49:131–136. doi:10.1016/j.etap.2016.11.019pl
dc.description.referencesPugazhendhi A, Edison TNJI, Karuppusamy I, Kathirvel B. Inorganic nanoparticles: a potential cancer therapy for human welfare. Int J Pharm. 2018;539(1–2):104–111. doi:10.1016/j.ijpharm. 2018.01.034pl
dc.description.referencesVasantharaj S, Sathiyavimal S, Senthilkumar P, LewisOscar F, Pugazhendhi A. Biosynthesis of iron oxide nanoparticles using leaf extract of Ruellia tuberosa: antimicrobial properties and their applications in photocatalytic degradation. J Photochem Photobiol B. 2019;192:74–82. doi:10.1016/j.jphotobiol.2018.12.025pl
dc.description.referencesJiang Z, Shan K, Song J, et al. Toxic effects of magnetic nanoparticles on normal cells and organs. Life Sci. 2019;220:156–161. doi:10.1016/j.lfs.2019.01.056pl
dc.description.referencesMalvindi MA, De Matteis V, Galeone A, et al. Toxicity assessment of silica coated iron oxide nanoparticles and biocompatibility improvement by surface engineering. PLoS One. 2014;9(1):e85835. doi:10.1371/journal.pone.0085835pl
dc.description.referencesNiemirowicz K, Piktel E, Wilczewska AZ, et al. Core-shell magnetic nanoparticles display synergistic antibacterial effects against Pseudomonas aeruginosa and Staphylococcus aureus when combined with cathelicidin LL-37 or selected ceragenins. Int J Nanomedicine. 2016;11:5443–5455. doi:10.2147/IJN.S113706pl
dc.description.referencesSurel U, Niemirowicz K, Marzec M, Savage PB, Bucki R. Ceragenins – a new weapon to fight multidrug resistant bacterial infections. Studia Medyczne. 2014;30(3):207–213. doi:10.5114/ ms.2014.45428pl
dc.description.referencesDing B, Guan Q, Walsh JP, et al. Correlation of the antibacterial activities of cationic peptide antibiotics and cationic steroid antibiotics. J Med Chem. 2002;45(3):663–669. doi:10.1021/jm0105070pl
dc.description.referencesMarkiewicz K, Zembko P, Półtorak K, et al. Magnetic nanoparticles with chelating shells prepared by RAFT/MADIX polymerization. New J Chem. 2016;40:9223–9231. doi:10.1039/C6NJ01938Bpl
dc.description.referencesWilczewska AZ, Markiewicz KH. Surface-Initiated RAFT/MADIX Polymerization on Xanthate-Coated Iron Oxide Nanoparticles. Macromol Chem Phys. 2014;215(2):190–197.pl
dc.description.referencesChou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58(3):621–681. doi:10.1124/pr.58.3.10pl
dc.description.referencesNiemirowicz K, Durnaś B, Tokajuk G, et al. Formulation and candidacidal activity of magnetic nanoparticles coated with cathelicidin LL-37 and ceragenin CSA-13. Sci Rep. 2017;7(1):4610. doi:10.1038/s41598-017-04653-1pl
dc.description.referencesSosa Iglesias V, Giuranno L, Dubois LJ, Theys J, Vooijs M. Drug resistance in non-small cell lung cancer: a potential for NOTCH targeting? Front Oncol. 2018;8:267. doi:10.3389/fonc.2018.00267pl
dc.description.referencesVan der Jeught K, Xu HC, Li YJ, Lu XB, Ji G. Drug resistance and new therapies in colorectal cancer. World J Gastroenterol. 2018;24 (34):3834–3848. doi:10.3748/wjg.v24.i34.3834pl
dc.description.referencesWnorowska U, Fiedoruk K, Piktel E, et al. Nanoantibiotics containing membrane-active human cathelicidin LL-37 or synthetic ceragenins attached to the surface of magnetic nanoparticles as novel and innovative therapeutic tools: current status and potential future applications. J Nanobiotechnol. 2020;18(1):3. doi:10.1186/s12951-019-0566-zpl
dc.description.referencesCampos SM, Penson RT, Mays AR, et al. The clinical utility of liposomal doxorubicin in recurrent ovarian cancer. Gynecol Oncol. 2001;81(2):206–212. doi:10.1006/gyno.2000.5980pl
dc.description.referencesKhan MA, Zafaryab M, Mehdi SH, Ahmad I, Rizvi MM. Characterization and anti-proliferative activity of curcumin loaded chitosan nanoparticles in cervical cancer. Int J Biol Macromol. 2016;93(Pt A):242–253. doi:10.1016/j.ijbiomac.2016.08.050pl
dc.description.referencesKuruvilla SP, Tiruchinapally G, Crouch AC, ElSayed MEH, Greve JM. Dendrimer-doxorubicin conjugates exhibit improved anticancer activity and reduce doxorubicin-induced cardiotoxicity in a murine hepatocellular carcinoma model. PLoS One. 2017;12(8): e0181944. doi:10.1371/journal.pone.0181944pl
dc.description.referencesPrylutska S, Grynyuk I, Matyshevska O, et al. C60 fullerene as synergistic agent in tumor-inhibitory Doxorubicin treatment. Drugs R D. 2014;14(4):333–340. doi:10.1007/s40268-014-0074-4pl
dc.description.referencesHekmat A, Saboury AA, Divsalar A. The effects of silver nanoparticles and doxorubicin combination on DNA structure and its antiproliferative effect against T47D and MCF7 cell lines. J Biomed Nanotechnol. 2012;8(6):968–982. doi:10.1166/jbn.2012.1451pl
dc.description.referencesDurnaś B, Piktel E, Wątek M, et al. Anaerobic bacteria growth in the presence of cathelicidin LL-37 and selected ceragenins delivered as magnetic nanoparticles cargo. BMC Microbiol. 2017;17(1):167. doi:10.1186/s12866-017-1075-6pl
dc.description.referencesTao X, Gou J, Zhang Q, et al. Synergistic breast tumor cell killing achieved by intracellular co-delivery of doxorubicin and disulfiram via core-shell-corona nanoparticles. Biomater Sci. 2018;6 (7):1869–1881. doi:10.1039/C8BM00271Apl
dc.description.referencesKaruppaiah A, Siram K, Selvaraj D, Ramasamy M, Babu D, Sankar V. Synergistic and enhanced anticancer effect of a facile surface modified non-cytotoxic silver nanoparticle conjugated with gemcitabine in metastatic breast cancer cells. Mater Today Commun. 2019;23:100884.pl
dc.description.referencesZhang R, Wang X, Wu C, et al. Synergistic enhancement effect of magnetic nanoparticles on anticancer drug accumulation in cancer cells. Nanotechnology. 2006;17(14):3622–3626. doi:10.1088/0957-4484/17/14/043pl
dc.description.referencesYin PT, Pongkulapa T, Cho HY, et al. Overcoming chemoresistance in cancer via combined microRNA therapeutics with anticancer drugs using multifunctional magnetic core-shell nanoparticles. ACS Appl Mater Interfaces. 2018;10(32):26954–26963. doi:10.1021/acsami.8b09086pl
dc.description.referencesAlarifi S, Ali D, Alkahtani S, Alhader MS. Iron oxide nanoparticles induce oxidative stress, DNA damage, and caspase activation in the human breast cancer cell line. Biol Trace Elem Res. 2014;159(1–-3):416–424. doi:10.1007/s12011-014-9972-0pl
dc.description.referencesWu W, Chen B, Cheng J, et al. Biocompatibility of Fe3O4/DNR magnetic nanoparticles in the treatment of hematologic malignancies. Int J Nanomedicine. 2010;5:1079–1084. doi:10.2147/IJN.S15660pl
dc.description.referencesRuden S, Hilpert K, Berditsch M, Wadhwani P, Ulrich AS. Synergistic interaction between silver nanoparticles and membrane-permeabilizing antimicrobial peptides. Antimicrob Agents Chemother. 2009;53(8):3538–3540. doi:10.1128/AAC.01106-08pl
dc.description.volume15pl
dc.description.firstpage4573pl
dc.description.lastpage4589pl
dc.identifier.citation2International Journal of Nanomedicinepl
dc.identifier.orcid0000-0002-4531-7277-
dc.identifier.orcid0000-0002-4857-0489-
dc.identifier.orcidbrakorcid-
dc.identifier.orcidbrakorcid-
dc.identifier.orcidbrakorcid-
dc.identifier.orcid0000-0002-3311-7147-
dc.identifier.orcid0000-0002-3386-146X-
dc.identifier.orcidbrakorcid-
dc.identifier.orcid0000-0002-4642-6109-
dc.identifier.orcid0000-0001-7664-9226-
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