We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Chemically synthesized ciprofloxacin-PEG-FeO nanotherapeutic exhibits strong antibacterial and controlled cytotoxic effects

    Hussan

    Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan

    ,
    Sobia Nisa

    *Author for correspondence:

    E-mail Address: sobia@uoh.edu.pk

    Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan

    ,
    Syeda Asma Bano

    Department of Microbiology, University of Haripur, Khyber Pakhtunkhwa, 22620, Pakistan

    &
    Muhammad Zia

    Department of Biotechnology, Quaid e Azam University Islamabad, Islamabad, 15320, Pakistan

    Published Online:https://doi.org/10.2217/nnm-2023-0298

    Aim: To develop a biocompatible conjugated ciprofloxacin-PEG-FeO nanodelivery system with increased efficacy of available therapeutics in a controlled manner. Materials & methods: FeO nanoparticles were synthesized by chemical and biological methods and modified as ciprofloxacin-PEG-FeO nanoformulations. After initial antibacterial and cytotoxicity studies, the effective and biocompatible nanoformulations was further fabricated as nanotherapeutics for in vivo studies in mouse models. Results: Chemically synthesized ciprofloxacin-PEG-FeO nanoformulations demonstrated boosted antibacterial activity against clinically isolated bacterial strains. Nanoformulations were also found to be compatible with baby hamster kidney 21 cells and red blood cells. In in vivo studies, nanotherapeutic showed wound-healing effects with eradication of Staphylococcus aureus infection. Conclusion: The investigations indicate that the developed nanotherapeutic can eradicate localized infections and enhance wound healing with controlled cytotoxicity.

    Tweetable abstract

    A ciprofloxacin-PEG-FeO nanodrug delivery system was developed, which showed enhanced antibiotic efficacy with a broad range of effects and controlled cytotoxicity in in vitro and in vivo experiments.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Parisi OI, Scrivano L, Sinicropi MS, Puoci F. Polymeric nanoparticle constructs as devices for antibacterial therapy. Curr. Opin. Pharmacol. 36, 72–77 (2017).
    • 2. Baranwal A, Srivastava A, Kumar P et al. Prospects of nano structure materials and their composites as antimicrobial agents. Front. Microbiol. 9, 422 (2018).
    • 3. Ashik UP, Kudo S, Hayashi JI. An overview of metal oxide nanostructures. In: Synthesis of Inorganic Nanomaterials: Advances and Key Technologies. Elsevier. 19–57 (2018).
    • 4. Martin-Serrano A, Gomez R, Ortega P, Mata FJ. Nanosystems as vehicles for the delivery of antimicrobial peptides (AMPs). Pharmaceutics 11, 448 (2019).
    • 5. Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug Discov. 9, 615 (2010).
    • 6. Pati R, Mehta RK, Mohanty S et al. Topical application of zinc oxide nanoparticles reduces bacterial skin infection in mice and exhibits antibacterial activity by inducing oxidative stress response and cell membrane disintegration in macrophages. Nanomedicine 10, 1195–1208 (2014).
    • 7. Premanathan M, Karthikeyan K, Jeyasubramanian K, Manivannan G. Selective toxicity of ZnO nanoparticles toward Gram-positive bacteria and cancer cells by apoptosis through lipid peroxidation. Nanomedicine 7, 184–192 (2011).
    • 8. Suka JS, Xua Q, Kima N, Hanesa J, Ensigna LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 99, 28–51 (2016).
    • 9. Vijeth S, Heggannavar GB, Kariduraganavar MY. Encapsulating wall materials for micro-/nanocapsules. In: Microencapsulation: Processes, Technologies and Industrial Applications. IntechOpen. 1–9 (2019).
    • 10. Xi-Feng XI, Jiang XQ, Lei-Ji ZH. Surface modification of polyethylene glycol to resist nonspecific adsorption of proteins. Chinese J. Anal. Chem. 41(3), 445–453 (2013).
    • 11. Shen Z, Loe DT, Fisher A et al. Polymer stiffness governs template mediated self-assembly of liposome-like nanoparticles: simulation, theory and experiment. Nanoscale 11(42), 20179–20193 (2019).
    • 12. Rossi LM, Quach AD, Rosenzweig Z. Glucose oxidase-magnetite nanoparticle bioconjugate for glucose sensing. Anal. Bioanal. Chem. 380, 606–613 (2004).
    • 13. Kaushik A, Khan R, Solanki PR et al. Iron oxide nanoparticles-chitosan composite based glucose biosensor. Biosens. Bioelectron. 24(4), 676–683 (2008).
    • 14. Cao D, Hu N. Direct electron transfer between hemoglobin and pyrolytic graphite electrodes enhanced by Fe3O4 nanoparticles in their layer-by-layer self-assembly films. Biophys. Chem. 121(3), 209–217 (2006).
    • 15. Gong J, Lin X. Facilitated electron transfer of hemoglobin embedded in nanosized Fe3O4 matrix based on paraffin impregnated graphite electrode and electrochemical catalysis for trichloroacetic acid. J. Nanobiotechnol. 75(1), 51–57 (2003).
    • 16. Ziarani, Ghodsi M, Lashgari N et al. The role of hollow magnetic nanoparticles in drug delivery. RSC Adv. 9.43, 25094–25106 (2019).
    • 17. Howard D, Jay M, Dziubla D et al. PEGylation of nanocarrier drug delivery systems: state of the art. J. B Nanotechnol. 4(2), 133–148 (2008).
    • 18. Rivas, Claudia JM, Badri W et al. Nanoprecipitation process: From encapsulation to drug delivery. Int. J. Pharmaceut. 532(1), 66–81 (2017).
    • 19. Farahmandjou M, Soflaee F. Synthesis and characterization of α-Fe2O3 nanoparticles by simple co-precipitation method. Phys. Chem. Res. 3(3), 191–196 (2015).
    • 20. Rodríguez-Perez C, Quirantes-Pine R, Fernandez-Gutierrez A, Segura-Carretero A. Optimization of extraction method to obtain a phenolic compounds-rich extract from Moringa oleifera Lam leaves. Ind. Crops Prod. 66, 246–254 (2015).
    • 21. Yew YP, Shameli K, Miyake M et al. Green synthesis of magnetite (Fe3O4) nanoparticles using seaweed (Kappaphycus alvarezii) extract. Nanoscale Res. Lett. 11, 1–7 (2016).
    • 22. Demir A, Topkaya R, Baykal A. Green synthesis of superparamagnetic Fe3O4 nanoparticles with maltose: its magnetic investigation. Polyhedron 65, 282–287 (2013).
    • 23. Zamora-Justo JA, Abrica-González P, Vázquez-Martínez G et al. Polyethylene glycol-coated gold nanoparticles as DNA and atorvastatin delivery systems and cytotoxicity evaluation. J. Nanomater. 2019, 1–11 (2019).
    • 24. Mohsen E, El-Borady OM, Mohamed MB, Fahim IS. Synthesis and characterization of ciprofloxacin loaded silver nanoparticles and investigation of their antibacterial effect. J. Radiat. Res. Appl. Sci. 13(1), 416–425 (2020).
    • 25. Giuliano C, Patel CR, Kale-Pradhan PB. A guide to bacterial culture identification and results interpretation. P T 44(4), 192 (2020).
    • 26. Ansari SA, Oves M, Satar R et al. Antibacterial activity of iron oxide nanoparticles synthesized by co-precipitation technology against Bacillus cereus and Klebsiella pneumoniae. Pol. J. Chem. Technol. 19(4), 110–115 (2017).
    • 27. Klancnik A, Piskernik S, Jersek B, Mozina SS. Evaluation of diffusion and dilution methods to determine the antibacterial activity of plant extracts. J. Microbiol. Methods 81(2), 121–126 (2010).
    • 28. Owuama CI. Determination of minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) using a novel dilution tube method. Afr. J. Microbiol. Res. 11(23), 977–980 (2017).
    • 29. Parvekar P, Palaskar J, Metgud S, Maria R, Dutta S. The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of silver nanoparticles against Staphylococcus aureus. Biomater. Investig. Dent. 7(1), 105–109 (2020).
    • 30. Cruz CD, Shah S, Tammela P. Defining conditions for biofilm inhibition and eradication assays for Gram-positive clinical reference strains. BMC Microbiol. 18(1), 1–9 (2018).
    • 31. Dosler S, Karaaslan E. Inhibition and destruction of Pseudomonas aeruginosa biofilms by antibiotics and antimicrobial peptides. Peptides 62, 32–37 (2014).
    • 32. Khan BA, Ullah S, Khan MK, Alshahrani SM, Braga VA. Formulation and evaluation of Ocimum basilicum-based emulgel for wound healing using animal model. Saudi Pharm. J. 28(12), 1842–1850 (2020).
    • 33. Shahidi F, Zhong Y. Measurement of antioxidant activity. J. Funct. Foods 18, 757–781 (2015).
    • 34. Sirivibulkovit K, Nouanthavong S, Sameenoi Y. Paper-based DPPH assay for antioxidant activity analysis. Anal. Sci. 34(7), 795–800 (2018).
    • 35. Tiwari V, Mishra N, Gadani K, Solanki PS, Shah NA, Tiwari M. Mechanism of anti-bacterial activity of zinc oxide nanoparticle against carbapenem-resistant Acinetobacter baumannii. Front. Microbiol. 9, 1218 (2018).
    • 36. Yang Z, Liu L, Su L et al. Design of a zero-order sustained release PLGA microspheres for palonosetron hydrochloride with high encapsulation efficiency. Int. J. Pharm. 575, 119006 (2020).
    • 37. Kumar P, Nagarajan A, Uchil PD. Analysis of cell viability by the MTT assay. Cold Spring Harb. Protoc. 2018(6), 095505 (2018).
    • 38. Fluhr JW, Darlenski R, Surber CJ. Glycerol and the skin: holistic approach to its origin and functions. Br. J. Dermatol. 159(1), 23–34 (2018).
    • 39. Long Z, Quaife B, Salman H, Oltvai ZN. Cell–cell communication enhances bacterial chemotaxis toward external attractants. Sci. Rep. 7(1), 12855 (2017).
    • 40. Kayani ZN, Arshad S, Riaz S, Naseem S. Synthesis of iron oxide nanoparticles by sol-gel technique and their characterization. IEEE Trans. Magn. 50(8), 1–4 (2014).
    • 41. Pu Y, Lu J, Wang D et al. Nanonization of ciprofloxacin using subcritical water-ethanol mixture as the solvent: solubility and precipitation parameters. Powder Technol. 321, 197–203 (2017).
    • 42. Aisida SO, Ugwu K, Akpa PA et al. Synthesis and characterization of iron oxide nanoparticles capped with Moringa oleifera: the mechanisms of formation effects on the optical, structural, magnetic and morphological properties. Mater. Today Proc. 36, 214–218 (2021).
    • 43. Vishalatchi M, Kalaiselvi V, Yasotha P et al. Eco-friendly synthesis of ferric oxide nanoparticles-antimicrobial activity. J. Environ. Nanotechnol. 11(3), 28–34 (2022).
    • 44. Aisida SO, Madubuonu N, Alnasir MH et al. Biogenic synthesis of iron oxide nanorods using Moringa oleifera leaf extract for antibacterial applications. Appl. Nanosci. 10, 305–315 (2020).
    • 45. Karpagavinayagam P, Vedhi C. Green synthesis of iron oxide nanoparticles using Avicennia marina flower extract. Vacuum 160, 286–292 (2019).
    • 46. Bharathi D, Ranjithkumar R, Vasantharaj S, Chandarshekar B, Bhuvaneshwari V. Synthesis and characterization of chitosan/iron oxide nanocomposite for biomedical applications. Int. J. Biol. Macromol. 132, 880–887 (2019).
    • 47. Sirivisoot S, Harrison BS. Magnetically stimulated ciprofloxacin release from polymeric microspheres entrapping iron oxide nanoparticles. Int. J. Nanomed. 10, 4447–4458 (2015). • Ciprofloxacin (CIP) releasing time was enhanced after loading with polycaprolactone-capped FeO nanoparticles (NPs).
    • 48. Mannu R, Vaithinathan K, Velu N et al. Polyethylene glycol coated magnetic nanoparticles: hybrid nanofluid formulation, properties and drug delivery prospects. Nanomaterials (Basel) 11(2), 440 (2021). •• Doxorubicin (DOX)-PEG-coated Ni-FeO nanoformulation revealed 1.3% DOX release in per mint, the study indicated polymer capping causes a slow release of drug from NPs surface.
    • 49. Mohamed N, Hessen OE, Mohammed HS. Thermal stability, paramagnetic properties, morphology and antioxidant activity of iron oxide nanoparticles synthesized by chemical and green methods. Inorg. Chem. Commun. 128, 108572 (2021). • Chemically synthesized FeO NPs showed high antioxidant properties in some stages compared with green synthesized FeO NPs using curcumin, Aloe vera and Green tea extracts.
    • 50. Ghaffar N, Javad S, Farrukh MA et al. Metal nanoparticles assisted revival of streptomycin against MDRS Staphylococcus aureus. PLOS ONE 17(3), 0264588 (2022).
    • 51. Abid MA, Kadhim DA, Aziz WJ. Iron oxide nanoparticle synthesis using Trigonella and tomato extracts and their antibacterial activity. Mater. Technol. 37(8), 547–554 (2022).
    • 52. Abadi B, Hosseinalipour S, Nikzad S et al. Capping agents for selenium nanoparticles in biomedical applications. J. Cluster Sci. 34(4), 1669–1690 (2023).
    • 53. Tortella G, Rubilar O, Fincheira P et al. Bactericidal and virucidal activities of biogenic metal-based nanoparticles: advances and perspectives. Antibiotics (Basel) 10(7), 783 (2021).
    • 54. Ali SA, Mmuo CC, Abdulraheem RO et al. High performance liquid chromatography (HPLC) method development and validation indicating assay for ciprofloxacin hydrochloride. J. Appl. Pharm. Sci. 1(8), 239–243 (2011).
    • 55. Billah MM, Rana SM, Hossain MS et al. Determination of the presence and pharmacokinetic profile of ciprofloxacin by TLC and HPLC method respectively in broiler chicken after single oral administration. J. Antibiot. (Tokyo) 67(11), 745–748 (2014).
    • 56. Arias LS, Pessan JP, Vieira AP et al. Iron oxide nanoparticles for biomedical applications: a perspective on synthesis, drugs, antimicrobial activity, and toxicity. Antibiotics (Basel) 7(2), 46 (2018). •• FeO NPs are biocompatible compared with many other metallic oxide NPs.
    • 57. Stoimenov PK, Klinger RL, Marchin GL, Klabunde KJ. Metal oxide nanoparticles as bactericidal agents. Langmuir 18(17), 6679–6686 (2002).
    • 58. Chen CZ, Cooper SL. Interactions between dendrimer biocides and bacterial membranes. Biomaterials 23(16), 3359–3368 (2002).
    • 59. SaodW, Al-Janaby MS, Gayadh EW et al. Biogenic synthesis of iron oxide nanoparticles using Hibiscus sabdariffa extract: potential for antibiotic development and antibacterial activity against multidrug-resistant bacteria. C.R.G Sustainable Chem. 8, 100397 (2024).
    • 60. Pedroso S, Seidy, Noralvis FS. The use of capping agents in the stabilization and functionalization of metallic nanoparticles for biomedical applications. P.P.S.C. 40(2), 2200146 (2023).
    • 61. Zhang S, Kucharski C, Doschak MR et al. Polyethylenimine–PEG coated albumin nanoparticles for BMP-2 delivery. Biomaterials 31(5), 952–963 (2010).
    • 62. Shirazi, Mehdi, Alireza A, Hadi S. Design and fabrication of magnetic Fe3O4-QSM nanoparticles loaded with ciprofloxacin as a potential antibacterial agent. I.J.B.M. 241, 124517 (2023).
    • 63. Shi L, Jinqiu Z, Zhao M et al. Effects of polyethylene glycol on the surface of nanoparticles for targeted drug delivery. Nanoscale 13(24), 10748–10764 (2021). • PEG capping on NPs can facilitate biocompatibility and drug delivery in a controlled manner.
    • 64. Sathiyaseelan A, Saravanakumar K, Mariadoss AV, Wang MH. Antimicrobial and wound healing properties of FeO fabricated chitosan/PVA nanocomposite sponge. Antibiotics (Basel) 10(5), 524 (2021). •• Synthesized chitosan/polyvinyl alcohol (PVA)-coated FeO nanocomposites evaluated skin wound-healing effects and its biocompatibility with kidney cell.
    • 65. Yudaev P, Mezhuev Y, Chistyakov E. Nanoparticle-containing wound dressing: antimicrobial and healing effects. Gels 8(6), 329 (2022). • An NP-based biocompatible delivery system can be useful in wound healing with antibacterial effects.