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

Modulating liposomal nanoparticles to enhance uptake and targeting of methicillin-resistant Staphylococcus aureus

    Mona Elhabak

    Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt

    ,
    Rania Ibrahim Shebl

    *Author for correspondence: Tel.: +202 023 833 3078;

    E-mail Address: Shbel.rania@gmail.com

    Department of Microbiology & Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt

    &
    Samia Omar

    Department of Pharmaceutical Technology, Faculty of Pharmacy, Horus University, New Domiate, Egypt

    Published Online:https://doi.org/10.2217/fmb-2022-0069

    Aims: To explore the role of modifying the phospholipid composition of liposomal nanoparticles (LNPs) on their uptake. Methods: Different LNPs were labeled with a fluorescent marker and their uptake by human lung fibroblast (WI-38) cells was evaluated using flow cytometry and confocal microscopy. Linezolid was loaded in LNPs showing enhanced uptake, and their ability to reduce intracellular methicillin-resistant Staphylococcus aureus (MRSA) was investigated by in vitro infection. Results: Liposomes with disaturated dipalmitoylphosphatidylcholine–phosphatidylglycerol–phosphatidylethanolamine at a molar ratio of 60:10:10, mimicking that of WI-38 cells, were more effectively uptaken. Linezolid-loaded LNPs significantly reduced intracellular MRSA viable count. Conclusion: Modified LNPs could be promising antibiotic nanocarriers for targeting intracellular MRSA, which are usually resistant to conventional antibiotics.

    Plain language summary

    Liposomal nanoparticles (LNPs) are considered effective drug-delivery nanocarriers. We investigated the effect of altering the phospholipid composition of LNPs on their uptake into lung cells. Intracellular uptake of LNPs with different phospholipids was evaluated. LNPs showing enhanced uptake were loaded with linezolid antibiotic and their potential to kill the intracellular bacteria was explored as the difficulty for an antibiotic to reach the intracellular bacteria results in treatment failure. LNPs with phospholipid composition similar to that of the lung cells were effectively uptaken and were also able to deliver linezolid into lung cells and kill the intracellular bacteria. This approach could be successfully applied to reduce the antibiotic dose and subsequently overcome antibiotic resistance.

    Tweetable abstract

    Liposomal nanocarriers with phospholipids mimicking that of lung cells could be an attractive approach in potentiating cellular uptake and reducing the viable count of intracellular methicillin-resistant Staphylococcus aureus.

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

    References

    • 1. Peters RJ, Bouwmeester H, Gottardo S et al. Nanomaterials for products and application in agriculture, feed and food. Trends Food Sci. Technol. 54, 155–164 (2016).
    • 2. Truong NP, Whittaker MR, Mak CW, Davis TP. The importance of nanoparticle shape in cancer drug delivery. Expert Opin. Drug Deliv. 12(1), 129–142 (2015).
    • 3. Ma N, Ma C, Li C et al. Influence of nanoparticle shape, size, and surface functionalization on cellular uptake. J. Nanosci. Nanotechnol. 13(10), 6485–6498 (2013). • Illustrates factors influencing the cellular uptake of liposomal nanoparticles.
    • 4. Alberts B, Johnson A, Lewis J et al. The lipid bilayer. In: Molecular Biology of the Cell (4th Edition). Garland Science, NY, USA (2002).
    • 5. Abdelkhaliq A, Van Der Zande M, Punt A et al. Impact of nanoparticle surface functionalization on the protein corona and cellular adhesion, uptake and transport. J. Nanobiotechnol. 16(1), 1–13 (2018).
    • 6. Dahal M, Schwan WR. Management of methicillin-resistant Staphylococcus aureus mediated ventilator-associated pneumonia. Curr. Trends Microbiol. 12, 95 (2018).
    • 7. Kishore S, Verma D, Siddique M. Comparison of in-vitro activities of linezolid and vancomycin against Staphylococcus aureus isolated from a tertiary care hospital. J. Clin. Diagn. Res. 8(5), DC12 (2014).
    • 8. Hajiahmadi F, Alikhani MY, Shariatifar H et al. The bactericidal effect of liposomal vancomycin as a topical combating system against methicillin-resistant Staphylococcus aureus skin wound infection in mice. Med. J. Islam. Repub. Iran 33, 153 (2019).
    • 9. Rivera AM, Boucher HW. Current concepts in antimicrobial therapy against select Gram-positive organisms: methicillin-resistant Staphylococcus aureus, penicillin-resistant pneumococci, and vancomycin-resistant enterococci. Mayo Clin. Proc. 86(12) 1230–1243 (2011). •• Describes the reason of linezolid selection to be loaded in the prepared liposomal nanoparticles with enhanced uptake efficiency.
    • 10. Ellington JK, Harris M, Hudson MC et al. Intracellular Staphylococcus aureus and antibiotic resistance: implications for treatment of staphylococcal osteomyelitis. J. Orthopaed. Res. 24(1), 87–93 (2006). •• Describes the importance of MRSA selection as an intracellular pathogen and the failure of treatment due to the low bioavailability of conventional antibiotics.
    • 11. Mohamed MF, Abdelkhalek A, Seleem MN. Evaluation of short synthetic antimicrobial peptides for treatment of drug-resistant and intracellular Staphylococcus aureus. Sci. Rep. 6(1), 1–14 (2016).
    • 12. Joyce P, Ulmefors H, Maghrebi S et al. Enhancing the cellular uptake and antibacterial activity of rifampicin through encapsulation in mesoporous silica nanoparticles. Nanomaterials 10(4), 815 (2020).
    • 13. Bashmail HA, Alamoudi AA, Noorwali A et al. Thymoquinone synergizes gemcitabine anti-breast cancer activity via modulating its apoptotic and autophagic activities. Sci. Rep. 8(1), 1–11 (2018).
    • 14. Dilnessa T, Bitew A. Prevalence and antimicrobial susceptibility pattern of methicillin resistant Staphylococcus aureus isolated from clinical samples at Yekatit 12 Hospital Medical College, Addis Ababa, Ethiopia. BMC Infect. Dis. 16(1), 1–9 (2016).
    • 15. Wayne P. Clinical and laboratory standards institute. Performance standards for antimicrobial susceptibility testing (2011). https://clsi.org/standards/products/microbiology/documents/m100/
    • 16. Haeri A, Alinaghian B, Daeihamed M, Dadashzadeh S. Preparation and characterization of stable nanoliposomal formulation of fluoxetine as a potential adjuvant therapy for drug-resistant tumors. Iranian J. Pharm. Res. 13(Suppl.), 3 (2014).
    • 17. Nogueira E, Cruz CF, Loureiro A et al. Assessment of liposome disruption to quantify drug delivery in vitro. Biochim. Biophys. Acta Biomembranes 1858(2), 163–167 (2016).
    • 18. Li X, Chen D, Le C et al. Novel mucus-penetrating liposomes as a potential oral drug-delivery system: preparation, in vitro characterization, and enhanced cellular uptake. Int. J. Nanomed. 6, 3151 (2011).
    • 19. Elmeshad AN, Mortazavi S, Mozafari MR. Formulation and characterization of nanoliposomal 5-fluorouracil for cancer nanotherapy. J. Liposome Res. 24(1), 1–9 (2014). • Evaluation of linezolid loading efficiency and capacity in the prepared liposomal nanoparticles.
    • 20. Farkas A, Pap B, Kondorosi É, Maróti G. Antimicrobial activity of NCR plant peptides strongly depends on the test assays. Front. Microbiol. 9, 2600 (2018).
    • 21. Shebl RI, Farouk F, Azzazy HME-S. Effect of surface charge and hydrophobicity modulation on the antibacterial and antibiofilm potential of magnetic iron nanoparticles. J. Nanomater. 2017, 3528295 (2017).
    • 22. Rollin G, Tan X, Tros F et al. Intracellular survival of Staphylococcus aureus in endothelial cells: a matter of growth or persistence. Front. Microbiol. 8, 1354 (2017). •• Important in performing the in vitro infection assay.
    • 23. Cheng C-Y, Olijve LL, Kausik R, Han S. Cholesterol enhances surface water diffusion of phospholipid bilayers. J. Chem. Phys. 141(22), 12B613_611 (2014).
    • 24. Danaei M, Dehghankhold M, Ataei S et al. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics 10(2), 57 (2018).
    • 25. Umbarkar M, Thakare S, Surushe T et al. Formulation and evaluation of liposome by thin film hydration method. J. Drug Deliv. Ther. 11(1), 72–76 (2021).
    • 26. Sou K. Electrostatics of carboxylated anionic vesicles for improving entrapment capacity. Chem. Phys. Lipids 164(3), 211–215 (2011).
    • 27. Yandrapati RK. Effect of lipid composition on the physical properties of liposomes: a light scattering study (2012). https://scholarsmine.mst.edu/masters_theses/6864/
    • 28. Yu K, Zhao J, Yu C et al. Role of four different kinds of polyethylenimines (PEIs) in preparation of polymeric lipid nanoparticles and their anticancer activity study. J. Cancer 7(7), 872 (2016). • Could justify difference between the zeta potentials of the two formulae.
    • 29. Makino K, Yamada T, Kimura M et al. Temperature-and ionic strength-induced conformational changes in the lipid head group region of liposomes as suggested by zeta potential data. Biophys. Chem. 41(2), 175–183 (1991).
    • 30. Chibowski E, Szcześ A. Zeta potential and surface charge of DPPC and DOPC liposomes in the presence of PLC enzyme. Adsorption 22(4), 755–765 (2016).
    • 31. Smith MC, Crist RM, Clogston JD, Mcneil SE. Zeta potential: a case study of cationic, anionic, and neutral liposomes. Anal. Bioanal. Chem. 409(24), 5779–5787 (2017).
    • 32. Fisher AB. Lung lipid composition and surfactant biology. In: Comparative Biology of the Normal Lung (2nd Edition). Parent RA (Ed.). Elsevier, 423–466 (2015). •• Describes the lipid composition of human lung fibroblast cells.
    • 33. Dziura M, Mansour B, Dipasquale M et al. Simulated breathing: application of molecular dynamics simulations to pulmonary lung surfactant. Symmetry 13(7), 1259 (2021).
    • 34. Radiom M, Sarkis M, Brookes O et al. Pulmonary surfactant inhibition of nanoparticle uptake by alveolar epithelial cells. Sci. Rep. 10(1), 1–14 (2020).
    • 35. Zhang R, Qin X, Kong F et al. Improving cellular uptake of therapeutic entities through interaction with components of cell membrane. Drug Deliv. 26(1), 328–342 (2019).
    • 36. Elhissi AM, Dennison SR, Ahmed W et al. New delivery systems – liposomes for pulmonary delivery of antibacterial drugs. Novel Antimicrob. Agents Strat. 387–406 (2014).
    • 37. Beltrán-Gracia E, López-Camacho A, Higuera-Ciapara I et al. Nanomedicine review: clinical developments in liposomal applications. Cancer Nanotechnol. 10(1), 1–40 (2019).
    • 38. Kirchner M, Higgins DE. Inhibition of ROCK activity allows InlF-mediated invasion and increased virulence of Listeria monocytogenes. Mol. Microbiol. 68(3), 749–767 (2008).
    • 39. Rocha CL, Rucks EA, Vincent DM, Olson JC. Examination of the coordinate effects of Pseudomonas aeruginosa ExoS on Rac1. Infect. Immun. 73(9), 5458–5467 (2005).
    • 40. Guo P, Buttaro BA, Xue HY et al. Lipid-polymer hybrid nanoparticles carrying linezolid improve treatment of methicillin-resistant Staphylococcus aureus (MRSA) harbored inside bone cells and biofilms. Eur. J. Pharm. Biopharm. 151, 189–198 (2020).
    • 41. Ferreira M, Ogren M, Dias JN et al. Liposomes as antibiotic delivery systems: a promising nanotechnological strategy against antimicrobial resistance. Molecules 26(7), 2047 (2021). • Could justify the enhanced reduction in the bacterial viable count in case of linezolid-loaded liposomes compared to free linezolid at low concentrations.