48 research outputs found
Recombinant human plasma gelsolin reverses increased permeability of the blood-brain barrier induced by the spike protein of the SARS-CoV-2 virus.
BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced levels of pGSN. Therefore, using an in vitro system, we investigated whether pGSN could attenuate increased permeability of the blood-brain barrier (BBB) during its exposure to the portion of the SARS-CoV-2 spike protein containing the receptor binding domain (S1 subunit).
MATERIALS AND METHODS: Two- and three-dimensional models of the human BBB were constructed using the human cerebral microvascular endothelial cell line hCMEC/D3 and exposed to physiologically relevant shear stress to mimic perfusion in the central nervous system (CNS). Trans-endothelial electrical resistance (TEER) as well as immunostaining and Western blotting of tight junction (TJ) proteins assessed barrier integrity in the presence of the SARS-CoV-2 spike protein and pGSN. The IncuCyte Live Imaging system evaluated the motility of the endothelial cells. Magnetic bead-based ELISA was used to determine cytokine secretion. Additionally, quantitative real-time PCR (qRT-PCR) revealed gene expression of proteins from signaling pathways that are associated with the immune response.
RESULTS: pGSN reversed S1-induced BBB permeability in both 2D and 3D BBB models in the presence of shear stress. BBB models exposed to pGSN also exhibited attenuated pro-inflammatory signaling pathways (PI3K, AKT, MAPK, NF-κB), reduced cytokine secretion (IL-6, IL-8, TNF-α), and increased expression of proteins that form intercellular TJ (ZO-1, occludin, claudin-5).
CONCLUSION: Due to its anti-inflammatory and protective effects on the brain endothelium, pGSN has the potential to be an alternative therapeutic target for patients with severe SARS-CoV-2 infection, especially those suffering neurological complications of COVID-19
Bactericidal and immunomodulatory properties of magnetic nanoparticles functionalized by 1,4-dihydropyridines
Background: 1,4-Dihydropyridine (1,4-DHP) and its derivatives are well-known calcium channel blockers with antiarrhythmic and antihypertensive activities. These compounds exhibit pleiotropic effects including antimicrobial activities that rely on their positive charge and amphipathic nature. Use of magnetic nanoparticles (MNPs) as carriers of 1,4-DHP modulates their properties and enables improved formulations with higher efficacy and less toxicity. Methods: In this study, the antimicrobial and immunomodulatory activities of novel 1,4-DHP derivatives in free form and immobilized on MNPs were determined by evaluating pathogen outgrowth and proinflammatory cytokine release in experimental settings that involve incubation of various 1,4-DHPs with clinical isolates of bacteria or fungi as well as mammalian cell culture models. Results: Conventional immobilization of 1,4-DHP on aminosilane-coated MNPs markedly enhances their antimicrobial activity compared to nonimmobilized molecules, in part because of the higher affinity of these nanosystems for bacterial cell wall components in the presence of human body fluids. Conclusion: Optimized nanosystems are characterized by improved biocompatibility and higher anti-inflammatory properties that provide new opportunities for the therapy of infectious diseasesThis study was supported by the National Science Center, Poland, under grant UMO-2015/17/B/NZ6/03473 (to RB). In 2016, KN-L was awarded a fellowship from the Foundation for Polish Science (FNP). This study was conducted with the use of equipment purchased by the Medical University of Białystok as a part of the RPOWP 2007–2013 funding, Priority I, Axis 1.1, contract no UDA-RPPD.01.01.00-20-001/15-00 dated June 26, 2015.Robert Bucki: [email protected] Niemirowicz-Laskowska - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokKatarzyna Głuszek - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokEwelina Piktel - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokKarlis Pajuste - Laboratory of Membrane Active Compounds and β-Diketones, Latvian Institute of Organic Synthesis, Riga, LatviaBonita Durnaś - Department of Microbiology and Immunology, The Faculty of Health Sciences of the Jan Kochanowski University in KielceGrzegorz Król - Department of Microbiology and Immunology, The Faculty of Health Sciences of the Jan Kochanowski University in KielceAgnieszka Z. Wilczewska - Institute of Chemistry, University of BialystokPaul A Janmey - Department of Physiology, Institute for Medicine and Engineering, University of PennsylvaniaAiva Plotniece - Laboratory of Membrane Active Compounds and β-Diketones, Latvian Institute of Organic Synthesis, Riga, LatviaRobert Bucki - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokYang J, Jiang C, Yang J, Qian C, Fang D. A clean procedure for the synthesis of 1,4-dihydropyridines via Hantzsch reaction in water. Green Chem Lett Rev. 2013;6(3):262–267.Filipan-Litvić M, Litvić M, Cepanec I, Vinković V. Hantzsch synthesis of 2,6-dimethyl-3,5-dimethoxycarbonyl-4-(o-methoxyphenyl)-1,4-dihydropyridine; a novel cyclisation leading to an unusual formation of 1-amino-2-methoxy-carbonyl-3,5-bis(o-methoxyphenyl)-4-oxacyclohexan-1-ene. Molecules. 2007;12(11):2546–2558.Khedkar SA, Auti PB. 1, 4-Dihydropyridines: a class of pharmacologically important molecules. Mini Rev Med Chem. 2014;14(3):282–290.Yamagishi S, Nakamura K, Takenaka K, Matsui T, Inoue H. Pleiotropic effects of nifedipine on atherosclerosis. Curr Pharm Des. 2006;12(12): 1543–1547.Toma L, Stancu CS, Sanda GM, Sima AV. Anti-oxidant and antiinflammatory mechanisms of amlodipine action to improve endothelial cell dysfunction induced by irreversibly glycated LDL. Biochem Biophys Res Commun. 2011;411(1):202–207.Prasanthi G, Prasad KV, Bharathi K. Synthesis, anticonvulsant activity and molecular properties prediction of dialkyl 1-(di(ethoxycarbonyl) methyl)-2,6-dimethyl-4-substituted-1,4-dihydropyridine-3,5-dicarboxylates. Eur J Med Chem. 2014;73:97–104.Kumar RS, Idhayadhulla A, Abdul Nasser AJ, Selvin J. Synthesis and anticoagulant activity of a new series of 1,4-dihydropyridine derivatives. Eur J Med Chem. 2011;46(2):804–810.Kumar R, Idhayadhulla A, Abdul Nasser A, Selvin J. Synthesis and antimicrobial activity of a new series of 1,4-dihydropyridine derivatives. J Serb Chem Soc. 2011;76(1):1–11.Reimão JQ, Scotti MT, Tempone AG. Anti-leishmanial and antitrypanosomal activities of 1,4-dihydropyridines: in vitro evaluation and structure-activity relationship study. Bioorg Med Chem. 2010;18(22): 8044–8053.Sirisha K, Achaiah G, Reddy VM. Facile synthesis and antibacterial, antitubercular, and anticancer activities of novel 1,4-dihydropyridines. Arch Pharm (Weinheim). 2010;343(6):342–352.Zorniak M, Mitrega K, Krzeminski TF. [Derivatives of 1,4-dihydropyridines as “priviledged structures” and their pharmacological potential]. Kardiol Pol. 2011;69(suppl 3):100–103. Polish.Marciniec B, Ogrodowczyk M. Photo- and radiostability of some 1,4-dihydropyridine derivatives in the solid state. Acta Pol Pharm. 2003; 60(2):151–155.Zhang D, Flint O, Wang L, et al. Cytochrome P450 11A1 bioactivation of a kinase inhibitor in rats: use of radioprofiling, modulation of metabolism, and adrenocortical cell lines to evaluate adrenal toxicity. Chem Res Toxicol. 2012;25(3):556–571.Miri R, Javidnia K, Amirghofran Z, et al. Cytotoxic effect of some 1,4-dihydropyridine derivatives containing nitroimidazole moiety. Iran J Pharm Res. 2011;10(3):497–503.Niemirowicz 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.Niemirowicz K, Durnaś B, Tokajuk G, et al. Magnetic nanoparticles as a drug delivery system that enhance fungicidal activity of polyene antibiotics. Nanomedicine. 2016;12(8):2395–2404.Taylor E, Webster TJ. Reducing infections through nanotechnology and nanoparticles. Int J Nanomedicine. 2011;6:1463–1473.Niemirowicz K, Durnaś B, Piktel E, Bucki R. Development of antifungal therapies using nanomaterials. Nanomedicine (Lond). 2017;12(15): 1891–1905.Kidd TJ, Mills G, Sá-Pessoa J, et al. A Klebsiella pneumoniae antibiotic resistance mechanism that subdues host defences and promotes virulence. EMBO Mol Med. 2017;9(4):430–447.Logan LK, Weinstein RA. The epidemiology of carbapenem-resistant enterobacteriaceae: the impact and evolution of a global menace. J Infect Dis. 2017;215(suppl_1):S28–S36.Massart R. Preparation of aqueous magnetic liquids in alkaline and acidic media. IEEE Trans Magn. 1981;17(2):1247–1248.Niemirowicz K, Swiecicka I, Wilczewska AZ, et al. Gold-functionalized magnetic nanoparticles restrict growth of Pseudomonas aeruginosa. Int J Nanomedicine. 2014;9:2217–2224.Meletiadis J, Mouton JW, Meis JF, et al. Comparison of spectrophotometric and visual readings of NCCLS method and evaluation of a colorimetric method based on reduction of a soluble tetrazolium salt, 2,3-bis [2-methoxy-4-nitro-5-[(sulfenylamino) carbonyl]-2H-tetrazoliumhydroxide], for antifungal susceptibility testing of Aspergillus species. J Clin Microbiol. 2001;39(12):4256–4263.Yao Y, Ohko Y, Sekiguchi Y, Fujishima A, Kubota Y. Self-sterilization using silicone catheters coated with Ag and TiO2 nanocomposite thin film. J Biomed Mater Res B Appl Biomater. 2008;85(2):453–460.Hussein-Al-Ali SH, El Zowalaty ME, Hussein MZ, Ismail M, Webster TJ. Synthesis, characterization, controlled release, and antibacterial studies of a novel streptomycin chitosan magnetic nanoantibiotic. Int J Nanomedicine. 2014;9:549–557.Niemirowicz 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.Sridhar J, Kuriyan AE, Flynn HW, Miller D. Endophthalmitis caused by Pseudomonas aeruginosa: clinical features, antibiotic susceptibilities, and treatment outcomes. Retina. 2015;35(6):1101–1106.Barreras US, Méndez FT, Martínez RE, Valencia CS, Rodríguez PR, Rodríguez JP. Chitosan nanoparticles enhance the antibacterial activity of chlorhexidine in collagen membranes used for periapical guided tissue regeneration. Mater Sci Eng C Mater Biol Appl. 2016;58:1182–1187.Feliciani C, Gupta AK, Sauder DN. Keratinocytes and cytokine/growth factors. Crit Rev Oral Biol Med. 1996;7(4):300–318.Gloire G, Legrand-Poels S, Piette J. NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol. 2006;72(11): 1493–1505.Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev. 2010;74(3):417–433.Katz AM, Leach NM. Differential effects of 1,4-dihydropyridine calcium channel blockers: therapeutic implications. J Clin Pharmacol. 1987; 27(11):825–834.Vijesh AM, Isloor AM, Peethambar SK, Shivananda KN, Arulmoli T, Isloor NA. Hantzsch reaction: synthesis and characterization of some new 1,4-dihydropyridine derivatives as potent antimicrobial and antioxidant agents. Eur J Med Chem. 2011;46(11):5591–5597.Briede J, Stivrina M, Vigante B, Stoldere D, Duburs G. Acute effect of antidiabetic 1,4-dihydropyridine compound cerebrocrast on cardiac function and glucose metabolism in the isolated, perfused normal rat heart. Cell Biochem Funct. 2008;26(2):238–245.Núñez-Vergara LJ, Squella JA, Bollo-Dragnic S, et al. Nitro aryl 1,4-dihydropyridine derivatives: effects on Trypanosoma cruzi. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol. 1997;118(1): 105–111.Sirisha K, Bikshapathi D, Achaiah G, Reddy VM. Synthesis, antibacterial and antimycobacterial activities of some new 4-aryl/heteroaryl-2,6-dimethyl-3,5-bis-N-(aryl)-carbamoyl-1,4-dihydropyridines. Eur J Med Chem. 2011;46(5):1564–1571.Chhillar AK, Arya P, Mukherjee C, et al. Microwave-assisted synthesis of antimicrobial dihydropyridines and tetrahydropyrimidin-2-ones: novel compounds against aspergillosis. Bioorg Med Chem. 2006; 14(4):973–981.Tasaka S, Ohmori H, Gomi N, et al. Synthesis and structure – activity analysis of novel dihydropyridine derivatives to overcome multidrug resistance. Bioorg Med Chem Lett. 2001;11(2):275–277.Miri R, Mehdipour A. Dihydropyridines and atypical MDR: a novel perspective of designing general reversal agents for both typical and atypical MDR. Bioorg Med Chem. 2008;16(18):8329–8334.Hetzel D, Strauss W, Bernard K, Li Z, Urboniene A, Allen LF. A phase III, randomized, open-label trial of ferumoxytol compared with iron sucrose for the treatment of iron deficiency anemia in patients with a history of unsatisfactory oral iron therapy. Am J Hematol. 2014;89(6): 646–650.Niemirowicz K, Markiewicz KH, Wilczewska AZ, Car H. Magnetic nanoparticles as new diagnostic tools in medicine. Adv Med Sci. 2012; 57(2):196–207.Piktel E, Niemirowicz K, Wątek M, Wollny T, Deptuła P, Bucki R. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnology. 2016;14(1):39.Azam A, Ahmed AS, Oves M, Khan MS, Habib SS, Memic A. Antimicrobial activity of metal oxide nanoparticles against Gram-positive and Gram-negative bacteria: a comparative study. Int J Nanomedicine. 2012;7:6003–6009.Leuba KD, Durmus NG, Taylor EN, Webster TJ. Short communication: carboxylate functionalized superparamagnetic iron oxide nanoparticles (SPION) for the reduction of S. aureus growth post biofilm formation. Int J Nanomedicine. 2013;8:731–736.Akbarzadeh T, Fallah Tafti A, Samadi N, et al. Synthesis and cloxacillin antimicrobial enhancement of 2-methylsulfonylimidazolyl-1,4-dihydropyridine derivatives. Daru. 2010;18(2):118–123.Bucki R, Janmey P. Extracellular aggregation of polyelectrolytes escaped from the cell interior: mechanisms and physiological consequences. Curr Opin Colloid Interface Sci. 2016;26:84–89.Cieśluk M, Piktel E, Wątek M, et al. Neutrophil extracellular traps as the main source of eDNA. Med Stud. 2017;33(2):137–145.Ruden 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): 3538–3540.133411342
Inhibition of inflammatory response in human keratinocytes by magnetic nanoparticles functionalized with PBP10 peptide derived from the PIP2-binding site of human plasma gelsolin
Background: 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.This 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.Robert Bucki: [email protected] Piktel - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokUrszula Wnorowska - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokMateusz Cieśluk - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokPiotr Deptula - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokKatarzyna Pogoda - IInstitute of Nuclear Physics Polish Academy of SciencesIwona Misztalewska‑Turkowicz - Institute of Chemistry, University of BiałystokPaulina Paprocka - Department of Microbiology and Immunology, The Faculty of Medicine and Health Sciences of the Jan Kochanowski University in KielceKatarzyna Niemirowicz‑Laskowska - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokAgnieszka Z. Wilczewska - Institute of Chemistry, University of BiałystokPaul A. Janmey - Department of Physiology and Institute for Medicine and Engineering, University of PennsylvaniaRobert Bucki - Department of Microbiological and Nanobiomedical Engineering, Medical University of BialystokSalmon JK, Armstrong CA, Ansel JC. The skin as an immune organ. West J Med. 1994;160(2):146–52Barker JN, Mitra RS, Grifths CE, Dixit VM, Nickolof BJ. Keratinocytes as initiators of infammation. Lancet. 1991;337(8735):211–4.Karin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial infections to chronic infammation and cancer. Cell. 2006;124(4):823–35.Kim 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.Gutsmann 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.Pfalzgraf 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.Eckmann 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.Bucki 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):4936–44Bucki 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.Bucki 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.Wątek 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.Bucki 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.Piktel 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.Bucki 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.Fu 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.Li 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.Nikapitiya 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.Lappas CM. The immunomodulatory efects of titanium dioxide and silver nanoparticles. Food Chem Toxicol. 2015;85:78–83.Ma 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.Gatto 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.Prasad 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.Moyano 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):320–7Pereira 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.Gregory 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.Niemirowicz-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.Massart R. Preparation of aqueous magnetic liquids in alkaline and acidic media. IEEE Trans Magn. 1981;17(2):1247–8.Niemirowicz 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.Dehghani 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.Dong 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.Durnaś 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):e0157242Yoon 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.Schwaminger 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.Niemirowicz 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.Bucki 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.Laskin JD, Heck DE, Laskin DL. Multifunctional role of nitric oxide in infammation. Trends Endocrinol Metab. 1994;5(9):377–82.Young 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.Sticherling 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.Pi 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.Pi 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.Kaminska 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.Gustafsson A, Olin AI, Ljunggren L. LPS interactions with immobilized and soluble antimicrobial peptides. Scand J Clin Lab Invest. 2010;70(3):194–200.Scott 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.Witke 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.Christofdou-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.Yang 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.Ordija 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.Taylor E, Webster TJ. Reducing infections through nanotechnology and nanoparticles. Int J Nanomedicine. 2011;6:1463–73.Liu 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.Nishanth 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.Niemirowicz 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.Lee 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):1922–30Mohamed 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.Carretero 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.Fiedoruk 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.Matsuzaki 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.Scott MG, Gold MR, Hancock RE. Interaction of cationic peptides with lipoteichoic acid and Gram-positive bacteria. Infect Immun. 1999;67(12):6445–53.Wang 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.Marcatili 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.Lai Y, Gallo RL. Toll-like receptors in skin infections and infammatory diseases. Infect Disord Drug Targets. 2008;8(3):144–55.Arany 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.Goldsmith 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.Herman AG, Moncada S. Therapeutic potential of nitric oxide donors in the prevention and treatment of atherosclerosis. Eur Heart J. 2005;26(19):1945–55.Wong VW, Lerner E. Nitric oxide inhibition strategies. Future Sci OA. 2015. https://doi.org/10.4155/fso.15.35.Bécherel 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.Nakashima 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.Grange 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.Ding 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.Bruch-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.Pogoda 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.Leporatti 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.Roca-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.Meng 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.Byfeld 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):6402–9.1
Recombinant Human Plasma Gelsolin Stimulates Phagocytosis while Diminishing Excessive Inflammatory Responses in Mice with Pseudomonas aeruginosa Sepsis
Plasma gelsolin (pGSN) is a highly conserved abundant circulating protein, characterized by diverse immunomodulatory activities including macrophage activation and the ability to neutralize pro-inflammatory molecules produced by the host and pathogen. Using a murine model of Gram-negative sepsis initiated by the peritoneal instillation of Pseudomonas aeruginosa Xen 5, we observed a decrease in the tissue uptake of IRDye®800CW 2-deoxyglucose, an indicator of inflammation, and a decrease in bacterial growth from ascitic fluid in mice treated with intravenous recombinant human plasma gelsolin (pGSN) compared to the control vehicle. Pretreatment of the murine macrophage line RAW264.7 with pGSN, followed by addition of Pseudomonas aeruginosa Xen 5, resulted in a dose-dependent increase in the proportion of macrophages with internalized bacteria. This increased uptake was less pronounced when cells were pretreated with pGSN and then centrifuged to remove unbound pGSN before addition of bacteria to macrophages. These observations suggest that recombinant plasma gelsolin can modulate the inflammatory response while at the same time augmenting host antibacterial activity.This work was supported by the National Science Center, Poland under Grant: UMO-2015/17/B/NZ6/03473 (to RB), National Institutes of Health: GM111942 (to PAJ) and Medical University of Bialystok: SUB/1/DN/19/001/1162 (to RB), N/ST/MN/18/001/1162 (to MC). 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. 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.Ewelina Piktel: [email protected] Wnorowska: [email protected] Cieśluk: [email protected] Deptuła: [email protected] V. Prasad: [email protected] Król: [email protected] Durnaś: [email protected] Namiot: [email protected] H. Markiewicz: [email protected] Niemirowicz-Laskowska: [email protected] Z. Wilczewska: [email protected] A. Janmey: [email protected] Reszeć: [email protected] Bucki: [email protected] Piktel - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokUrszula Wnorowska - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokMateusz Cieśluk - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokPiotr Deptuła - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokSuhanya V. Prasad - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokGrzegorz Król - Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in KielceBonita Durnaś - Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in KielceAndrzej Namiot - Department of Anatomy, Medical University of BialystokKarolina H. Markiewicz - Institute of Chemistry, University of BiałystokKatarzyna Niemirowicz-Laskowska - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of BialystokAgnieszka Z. Wilczewska - Institute of Chemistry, University of BiałystokPaul A. Janmey - Institute for Medicine and Engineering, University of PennsylvaniaJoanna Reszeć - Department of Pathology, Medical University of BialystokRobert Bucki - Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok; Department of Microbiology and Immunology, the Faculty of Medicine and Health Sciences of the Jan Kochanowski University in KielceLee, P.S.; Patel, S.R.; Christiani, D.C.; Bajwa, E.; Stossel, T.P.; Waxman, A.B. Plasma gelsolin depletion and circulating actin in sepsis: A pilot study. PLoS ONE 2008, 3, e3712.Li-ChunHsieh, K.; Schob, S.; Zeller, M.W.; Pulli, B.; Ali, M.; Wang, C.; Chiou, T.T.; Tsang, Y.M.; Lee, P.S.; Stossel, T.P.; et al. Gelsolin decreases actin toxicity and inflammation in murine multiple sclerosis. J. Neuroimmunol. 2015, 287, 36–42.Bucki, R.; Kulakowska, A.; Byfield, F.J.; Zendzian-Piotrowska, M.; Baranowski, M.; Marzec, M.; Winer, J.P.; Ciccarelli, N.J.; Górski, J.; Drozdowski, W.; et al. Plasma gelsolin modulates cellular response to sphingosine 1-phosphate. Am. J. Physiol. Cell Physiol. 2010, 299, C1516–C1523.Bucki, R.; Georges, P.C.; Espinassous, Q.; Funaki, M.; Pastore, J.J.; Chaby, R.; Janmey, P.A. Inactivation of endotoxin by human plasma gelsolin. Biochemistry 2005, 44, 9590–9597.Piktel, E.; Levental, I.; Durnas, B.; Janmey, P.A.; Bucki, R. Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target. Int. J. Mol. Sci. 2018, 19, 2516.Christofidou-Solomidou, M.; Scherpereel, A.; Solomides, C.C.; Muzykantov, V.R.; Machtay, M.; Albelda, S.M.; DiNubile, M.J. Changes in plasma gelsolin concentration during acute oxidant lung injury in mice. Lung 2002, 180, 91–104.Xianhui, L.; Pinglian, L.; Xiaojuan, W.; Wei, C.; Yong, Y.; Feng, R.; Peng, S.; Gang, X. The association between plasma gelsolin level and prognosis of burn patients. Burns 2014, 40, 1552–1555.Wang, H.; Cheng, B.; Chen, Q.; Wu, S.; Lv, C.; Xie, G.; Jin, Y.; Fang, X. Time course of plasma gelsolin concentrations during severe sepsis in critically ill surgical patients. Crit. Care 2008, 12, R106.Marrocco, C.; Rinalducci, S.; Mohamadkhani, A.; D’Amici, G.M.; Zolla, L. Plasma gelsolin protein: A candidate biomarker for hepatitis B-associated liver cirrhosis identified by proteomic approach. Blood Transfus. 2010, 8, S105–S112.Stalmach, A.; Johnsson, H.; McInnes, I.B.; Husi, H.; Klein, J.; Dakna, M.; Mullen, W.; Mischak, H.; Porter, D. Identification of urinary peptide biomarkers associated with rheumatoid arthritis. PLoS ONE 2014, 9, e104625.Hu, Y.; Li, H.; Li, W.H.; Meng, H.X.; Fan, Y.Z.; Li, W.J.; Ji, Y.T.; Zhao, H.; Zhang, L.; Jin, X.M.; et al. The value of decreased plasma gelsolin levels in patients with systemic lupus erythematosus and rheumatoid arthritis in diagnosis and disease activity evaluation. Lupus 2013, 22, 1455–1461.Lee, P.S.; Waxman, A.B.; Cotich, K.L.; Chung, S.W.; Perrella, M.A.; Stossel, T.P. Plasma gelsolin is a marker and therapeutic agent in animal sepsis. Crit. Care Med. 2007, 35, 849–855.Bucki, R.; Levental, I.; Kulakowska, A.; Janmey, P.A. Plasma gelsolin: Function, prognostic value, and potential therapeutic use. Curr. Protein. Pept. Sci. 2008, 9, 541–551.Cohen, T.S.; Bucki, R.; Byfield, F.J.; Ciccarelli, N.J.; Rosenberg, B.; DiNubile, M.J.; Janmey, P.A.; Margulies, S.S. Therapeutic potential of plasma gelsolin administration in a rat model of sepsis. Cytokine 2011, 54, 235–238.Marshall, M.V.; Draney, D.; Sevick-Muraca, E.M.; Olive, D.M. Single-dose intravenous toxicity study of IRDye 800CW in Sprague-Dawley rats. Mol. Imaging Biol. 2010, 12, 583–594.Kovar, J.L.; Volcheck, W.; Sevick-Muraca, E.; Simpson, M.A.; Olive, D.M. Characterization and performance of a near-infrared 2-deoxyglucose optical imaging agent for mouse cancer models. Anal. Biochem. 2009, 384, 254–262.Nanda, J.S.; Lorsch, J.R. Labeling a protein with fluorophores using NHS ester derivitization. Methods Enzymol. 2014, 536, 87–94.Mounzer, K.C.; Moncure, M.; Smith, Y.R.; Dinubile, M.J. Relationship of admission plasma gelsolin levels to clinical outcomes in patients after major trauma. Am. J. Respir. Crit. Care Med. 1999, 160, 1673–1681.Osborn, T.M.; Verdrengh, M.; Stossel, T.P.; Tarkowski, A.; Bokarewa, M. Decreased levels of the gelsolin plasma isoform in patients with rheumatoid arthritis. Arthritis Res. Ther. 2008, 10, R117.Lee, P.S.; Sampath, K.; Karumanchi, S.A.; Tamez, H.; Bhan, I.; Isakova, T.; Gutierrez, O.M.; Wolf, M.; Chang, Y.; Stossel, T.P.; et al. Plasma gelsolin and circulating actin correlate with hemodialysis mortality. J. Am. Soc. Nephrol. 2009, 20, 1140–1148.Mochizuki, T.; Tsukamoto, E.; Kuge, Y.; Kanegae, K.; Zhao, S.; Hikosaka, K.; Hosokawa, M.; Kohanawa, M.; Tamaki, N. FDG uptake and glucose transporter subtype expressions in experimental tumor and inflammation models. J. Nucl. Med. 2001, 42, 1551–1555.Wittmann, J.; Dieckow, J.; Schröder, H.; Hampel, U.; Garreis, F.; Jacobi, C.; Milczarek, A.; Hsieh, K.L.; Pulli, B.; Chen, J.W.; et al. Plasma gelsolin promotes re-epithelialization. Sci. Rep. 2018, 8, 13140.Witke, W.; Sharpe, A.H.; Hartwig, J.H.; Azuma, T.; Stossel, T.P.; Kwiatkowski, D.J. Hemostatic, inflammatory, and fibroblast responses are blunted in mice lacking gelsolin. Cell 1995, 81, 41–51.Lind, S.E.; Smith, D.B.; Janmey, P.A.; Stossel, T.P. Role of plasma gelsolin and the vitamin D-binding protein in clearing actin from the circulation. J. Clin. Investig. 1986, 78, 736–742.Serrander, L.; Skarman, P.; Rasmussen, B.; Witke, W.; Lew, D.P.; Krause, K.H.; Stendahl, O.; Nüsse, O. Selective inhibition of IgG-mediated phagocytosis in gelsolin-deficient murine neutrophils. J. Immunol. 2000, 165, 2451–2457.Arora, P.D.; Glogauer, M.; Kapus, A.; Kwiatkowski, D.J.; McCulloch, C.A. Gelsolin mediates collagen phagocytosis through a rac-dependent step. Mol. Biol. Cell 2004, 15, 588–599.Jutras, I.; Desjardins, M. Phagocytosis: At the crossroads of innate and adaptive immunity. Annu. Rev. Cell Dev. Biol. 2005, 21, 511–527.Kitchens, R.L.; Thompson, P.A. Modulatory effects of sCD14 and LBP on LPS-host cell interactions. J. Endotoxin. Res. 2005, 11, 225–229.Lamping, N.; Dettmer, R.; Schröder, N.W.; Pfeil, D.; Hallatschek, W.; Burger, R.; Schumann, R.R. LPS-binding protein protects mice from septic shock caused by LPS or gram-negative bacteria. J. Clin. Investig. 1998, 101, 2065–2071.Jack, R.S.; Fan, X.; Bernheiden, M.; Rune, G.; Ehlers, M.; Weber, A.; Kirsch, G.; Mentel, R.; Fürll, B.; Freudenberg, M.; et al. Lipopolysaccharide-binding protein is required to combat a murine gram-negative bacterial infection. Nature 1997, 389, 742–745.Schiff, D.E.; Kline, L.; Soldau, K.; Lee, J.D.; Pugin, J.; Tobias, P.S.; Ulevitch, R.J. Phagocytosis of gram-negative bacteria by a unique CD14-dependent mechanism. J. Leukoc. Biol. 1997, 62, 786–794.Klein, R.D.; Su, G.L.; Schmidt, C.; Aminlari, A.; Steinstraesser, L.; Alarcon, W.H.; Zhang, H.Y.; Wang, S.C. Lipopolysaccharide-binding protein accelerates and augments Escherichia coli phagocytosis by alveolar macrophages. J. Surg. Res. 2000, 94, 159–166.Bucki, R.; Niemirowicz, K.; Wnorowska, U.; Byfield, F.J.; Piktel, E.; W ˛atek, M.; Janmey, P.A.; Savage, P.B. Bactericidal activity of ceragenin CSA-13 in cell culture and an animal model of peritoneal infection. Antimicrob. Agents Chemother. 2015.21711
Extracellular Vimentin as a Target Against SARS-CoV-2 Host Cell Invasion
Infection of human cells by pathogens, including SARS-CoV-2, typically proceeds by cell surface binding to a crucial receptor. The primary receptor for SARS-CoV-2 is the angiotensin-converting enzyme 2 (ACE2), yet new studies reveal the importance of additional extracellular co-receptors that mediate binding and host cell invasion by SARS-CoV-2. Vimentin is an intermediate filament protein that is increasingly recognized as being present on the extracellular surface of a subset of cell types, where it can bind to and facilitate pathogens’ cellular uptake. Biophysical and cell infection studies are done to determine whether vimentin might bind SARS-CoV-2 and facilitate its uptake. Dynamic light scattering shows that vimentin binds to pseudovirus coated with the SARS-CoV-2 spike protein, and antibodies against vimentin block in vitro SARS-CoV-2 pseudovirus infection of ACE2-expressing cells. The results are consistent with a model in which extracellular vimentin acts as a co-receptor for SARS-CoV-2 spike protein with a binding affinity less than that of the spike protein with ACE2. Extracellular vimentin may thus serve as a critical component of the SARS-CoV-2 spike protein-ACE2 complex in mediating SARS-CoV-2 cell entry, and vimentin-targeting agents may yield new therapeutic strategies for preventing and slowing SARS-CoV-2 infection
Bone Marrow Osteoblast Damage by Chemotherapeutic Agents
Hematopoietic reconstitution, following bone marrow or stem cell transplantation, requires a microenvironment niche capable of supporting both immature progenitors and stem cells with the capacity to differentiate and expand. Osteoblasts comprise one important component of this niche. We determined that treatment of human primary osteoblasts (HOB) with melphalan or VP-16 resulted in increased phospho-Smad2, consistent with increased TGF-β1 activity. This increase was coincident with reduced HOB capacity to support immature B lineage cell chemotaxis and adherence. The supportive deficit was not limited to committed progenitor cells, as human embryonic stem cells (hESC) or human CD34+ bone marrow cells co-cultured with HOB pre-exposed to melphalan, VP-16 or rTGF-β1 had profiles distinct from the same populations co-cultured with untreated HOB. Functional support deficits were downstream of changes in HOB gene expression profiles following chemotherapy exposure. Melphalan and VP-16 induced damage of HOB suggests vulnerability of this critical niche to therapeutic agents frequently utilized in pre-transplant regimens and suggests that dose escalated chemotherapy may contribute to post-transplantation hematopoietic deficits by damaging structural components of this supportive niche
Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target
Gelsolin, an actin-depolymerizing protein expressed both in extracellular fluids and in the cytoplasm of a majority of human cells, has been recently implicated in a variety of both physiological and pathological processes. Its extracellular isoform, called plasma gelsolin (pGSN), is present in blood, cerebrospinal fluid, milk, urine, and other extracellular fluids. This isoform has been recognized as a potential biomarker of inflammatory-associated medical conditions, allowing for the prediction of illness severity, recovery, efficacy of treatment, and clinical outcome. A compelling number of animal studies also demonstrate a broad spectrum of beneficial effects mediated by gelsolin, suggesting therapeutic utility for extracellular recombinant gelsolin. In the review, we summarize the current data related to the potential of pGSN as an inflammatory predictor and therapeutic target, discuss gelsolin-mediated mechanisms of action, and highlight recent progress in the clinical use of pGSN
Toxicity of parasites and their unconventional use in medicine
Introduction. Over 300 species of parasites can possibly be passed on humans. Most of the parasitic infections are defined based on their pathogenicity; however, some positive effects of a parasite existence within the human body have recently been suggested. Beneficial outcomes of parasite infections might result from the production and release of metabolites, modification of host immune response or products uptake of the host.
Objective. The aim of the study was a comprehensive analysis of a wide range of effects of parasites on the human body, including an overview of the toxic and positive effects.
State of knowledge. In the light of the latest research presenting the unconventional use of parasites in medicine, the widely understood of their impact on the human body can also be considered in a positive context. Clinical cases from diseases caused by the toxic effects of parasites, as described in recent years, indicate that the problem of parasitic infections still persists. Despite a great deal of knowledge about the toxic effects of parasites on the human organism and, above all, despite the improvement in sanitary conditions, there is a resurgence of parasitic infections, as evidenced, e.g. by the examples presented in this review.
Conclusions. The examples of positive effects of parasites presented so far give hope for the future in terms of fighting many diseases for which pharmacological treatment has not yet brought a positive effect. A better understanding of those processes might lead to the development of new methods of unconventional medical treatment