1887

Abstract

In the present study, enhancement of the the antibacterial activity of ceftriaxone against Gram-positive (meticillin-resistant ; MRSA) and Gram-negative () bacteria with a biodegradable polymer was attempted.

MRSA and were collected and identified by biochemical and molecular tests. Blank and ceftriaxone-loaded chitosan nanoparticles (CNPs) were prepared by the ionic gelation method. antibiotic-susceptibility studies were performed by disc diffusion, agar well plate method, Etest and time-kill assay. activity was assessed using the neutropenic mouse thigh model and cytotoxicity was estimated by MTT (methylthiazolyldiphenyl tetrazolium bromide) assay with the MCF-7 cancer cell line.

MRSA showed 97 % and 83 % resistance against ceftriaxone in the disc diffusion test. The isolates showing a ≥1024 mg l MIC value for ceftriaxone were selected for further evaluation. In the agar well plate method, the mean zones of inhibition for blank and ceftriaxone-loaded CNPs were 17 and 23 mm, respectively, for MRSA isolates and 15 and 25 mm, respectively, for isolates. In the time-kill assay, ~1 log to ~2.5 log reduction in viability was seen with both isolates when treated with ceftriaxone-loaded CNPs over 24 h. The studies also showed the enhanced antibacterial activity of ceftriaxone-loaded CNPs, with a 41 % reduction in MRSA and a 27 % reduction in burden. A low cytotoxicity of blank and ceftriaxone-loaded CNPs was seen, with a slight reduction in the percentage viability of cells from 87 to 83 % and from 88 to 81 %, respectively.

The synergistic effect of ceftriaxone-loaded CNPs is a useful finding for the treatment of MRSA and infections.

Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.000445
2017-03-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jmm/66/3/318.html?itemId=/content/journal/jmm/10.1099/jmm.0.000445&mimeType=html&fmt=ahah

References

  1. Blaser MJ. Who are we? Indigenous microbes and the ecology of human diseases. EMBO Rep 2006; 7:956–960 [View Article][PubMed]
    [Google Scholar]
  2. Diekema DJ, Pfaller MA, Schmitz FJ, Smayevsky J, Bell J et al. Survey of infections due to Staphylococcus species: frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, Latin America, Europe, and the Western Pacific region for the SENTRY Antimicrobial Surveillance Program, 19971999. Clin Infect Dis 2001; 32:S114–S132 [View Article][PubMed]
    [Google Scholar]
  3. Allocati N, Masulli M, Alexeyev MF, Di Ilio C. Escherichia coli in Europe: an overview. Int J Environ Res Public Health 2013; 10:6235–6254 [View Article][PubMed]
    [Google Scholar]
  4. Van der Bij AK, Pitout JD. The role of international travel in the worldwide spread of multiresistant Enterobacteriaceae. J Antimicrob Chemother 2012; 67:2090–2100 [View Article][PubMed]
    [Google Scholar]
  5. McEachran AD, Blackwell BR, Hanson JD, Wooten KJ, Mayer GD et al. Antibiotics, bacteria, and antibiotic resistance genes: aerial transport from cattle feed yards via particulate matter. Environ Health Perspect 2015; 123:337–343 [View Article][PubMed]
    [Google Scholar]
  6. Schmitz FJ, Jones ME. Antibiotics for treatment of infections caused by MRSA and elimination of MRSA carriage. What are the choices?. Int J Antimicrob Agents 1997; 9:1–19 [View Article][PubMed]
    [Google Scholar]
  7. Cui L, Tominaga E, Neoh HM, Hiramatsu K. Correlation between reduced daptomycin susceptibility and vancomycin resistance in vancomycin-intermediate Staphylococcus aureus. Antimicrob Agents Chemother 2006; 50:1079–1082 [View Article][PubMed]
    [Google Scholar]
  8. Collignon P. Resistant Escherichia coli — we are what we eat. Clin Infect Dis 2009; 49:202–204 [View Article][PubMed]
    [Google Scholar]
  9. Ali SR, Ahmed S, Lohana H. Trends of empiric antibiotic usage in a secondary care hospital, Karachi, Pakistan. Int J Pediatr 2013; 2013:1–4 [View Article]
    [Google Scholar]
  10. O'Callaghan KAM, Kerry JP. Preparation of low- and medium-molecular weight chitosan nanoparticles and their antimicrobial evaluation against a panel of microorganisms, including cheese-derived cultures. Food Control 2016; 69:256–261 [View Article]
    [Google Scholar]
  11. Tsai T, Chien HF, Wang TH, Huang CT, Ker YB et al. Chitosan augments photodynamic inactivation of Gram-positive and Gram-negative bacteria. Antimicrob Agents Chemother 2011; 55:1883–1890 [View Article][PubMed]
    [Google Scholar]
  12. Kong M, Chen XG, Xing K, Park HJ. Antimicrobial properties of chitosan and mode of action: a state of the art review. Int J Food Microbiol 2010; 144:51–63 [View Article][PubMed]
    [Google Scholar]
  13. Qi L, Xu Z, Jiang X, Hu C, Zou X. Preparation and antibacterial activity of chitosan nanoparticles. Carbohydr Res 2004; 339:2693–2700 [View Article][PubMed]
    [Google Scholar]
  14. Murakami K, Minamide W, Wada K, Nakamura E, Teraoka H et al. Identification of methicillin-resistant strains of staphylococci by polymerase chain reaction.. J Clin Microbiol 1991; 29:2240–2244[PubMed]
    [Google Scholar]
  15. Brakstad OG, Aasbakk K, Maeland JA. Detection of Staphylococcus aureus by polymerase chain reaction amplification of the nuc gene.. J Clin Microbiol 1992; 30:1654–1714[PubMed]
    [Google Scholar]
  16. Chen J, Griffiths MW. PCR differentiation of Escherichia coli from other gram-negative Bacteria using primers derived from the nucleotide sequences flanking the gene encoding the universal stress protein. Lett Appl Microbiol 1998; 27:369–371 [View Article][PubMed]
    [Google Scholar]
  17. European Committee on Antimicrobial Susceptibility Testing (EUCAST) 2015; Antimicrobial susceptibility testing of bacteria. http://www.eucast.org/ast_of_bacteria/
  18. Anitha A, Deepa N, Chennazhi KP, Nair SV, Tamura H et al. Development of mucoadhesive thiolated chitosan nanoparticles for biomedical applications. Carbohydr Polym 2011; 83:66–73 [View Article]
    [Google Scholar]
  19. Cordente N, Respaud M, Senocq F, Casanove M-J, Amiens C et al. Synthesis and magnetic properties of nickel nanorods. Nano Lett 2001; 1:565–568 [View Article]
    [Google Scholar]
  20. Song Y, Zhu A, Song Y, Cheng Z, Xu J et al. Experimental and theoretical study on the synthesis of gold nanoparticles using ceftriaxone as a stabilizing reagent for and its catalysis for dopamine. Gold Bull 2012; 45:153–160 [CrossRef]
    [Google Scholar]
  21. Jen SU, Cheng WC, Chiang FL. Structural, magneto-mechanical, and damping properties of slowly-cooled polycrystalline Fe81Ga19 alloy. J Alloys Compd 2015; 651:544–550 [View Article]
    [Google Scholar]
  22. Al-Zahrani IA, Hamson C, Edge D, Collins J, Perry JD et al. SmaI restriction site-based multiplex PCR for typing of hospital- and community-acquired Staphylococcus aureus. J Clin Microbiol 2011; 49:3820–3828 [View Article][PubMed]
    [Google Scholar]
  23. Osek J. Multiplex polymerase chain reaction assay for identification of enterotoxigenic Escherichia coli strains. J Vet Diagn Invest 2001; 13:308–311 [View Article][PubMed]
    [Google Scholar]
  24. Chitnis S, Katara G, Hemvani N, Pareek S, Chitnis DS. In vitro activity of daptomycin & linezolid against methicillin resistant Staphylococcus aureus & vancomycin resistant enterococci isolated from hospitalized cases in Central India. Indian J Med Res 2013; 137:191–196[PubMed]
    [Google Scholar]
  25. Sharma P, Vishwanath G. Study of vancomycin susceptibility in methicillin-resistant Staphylococcus aureus isolated from clinical samples. Ann Trop Med PH 2012; 5:178–180 [View Article]
    [Google Scholar]
  26. Moş I, Micle O, Zdrâncă M, Mureşan M, Vicaş L. Antibiotic sensitivity of the Escherichia coli strains isolated from infected skin wounds. Farmacia 2010; 58:637–645
    [Google Scholar]
  27. Phe K, Dao D, Palmer HR, Tam VH. In vitro ceftriaxone susceptibility in methicillin-susceptible Staphylococcus aureus. Antimicrob Agents Chemother 2015; 59:1370 [View Article][PubMed]
    [Google Scholar]
  28. Zakaria AS, Melake NA, Baky NA, El Rasheed NM, Ibrahim NH. In vitro and in vivo studies of antibacterial effect of ceftriaxone moxifloxacin combination against methicillin resistant Staphylococcus aureus biofilms formed on biomedical implants. Afr J Microbiol Res 2012; 6:5399–5409
    [Google Scholar]
  29. Hwang KP, Tang YF, Shen YH. Activity of ertapenem, ciprofloxacin, ceftriaxone, piperacillin-tazobactam, and ampicillin-sulbactam against 12 common clinical isolates of community-acquired bacteremia. J Microbiol Immunol Infect 2009; 42:433–438[PubMed]
    [Google Scholar]
  30. Du WL, Xu YL, Xu ZR, Fan CL. Preparation, characterization and antibacterial properties against E. coli K(88) of chitosan nanoparticle loaded copper ions. Nanotechnology 2008; 19:085707 [View Article][PubMed]
    [Google Scholar]
  31. Romero-Pérez A, García-García E, Zavaleta-Mancera A, Ramírez-Bribiesca JE, Revilla-Vázquez A et al. Designing and evaluation of sodium selenite nanoparticles in vitro to improve selenium absorption in ruminants. Vet Res Commun 2010; 34:71–79 [View Article][PubMed]
    [Google Scholar]
  32. Raula J, Eerikäinen H, Kauppinen EI. Influence of the solvent composition on the aerosol synthesis of pharmaceutical polymer nanoparticles. Int J Pharm 2004; 284:13–21 [View Article][PubMed]
    [Google Scholar]
  33. Unsoy G, Yalcin S, Khodadust R, Gunduz G, Gunduz U. Synthesis optimization and characterization of chitosan-coated iron oxide nanoparticles produced for biomedical applications. J Nanopart Res 2012; 14:964 [View Article]
    [Google Scholar]
  34. Thomas V, Yallapu MM, Sreedhar B, Bajpai SK. Fabrication, characterization of chitosan/nanosilver film and its potential antibacterial application. J Biomater Sci Polym Ed 2009; 20:2129–2144 [View Article][PubMed]
    [Google Scholar]
  35. Zaki NM, Hafez MM. Enhanced antibacterial effect of ceftriaxone sodium-loaded chitosan nanoparticles against intracellular Salmonella typhimurium. AAPS PharmSciTech 2012; 13:411–421 [View Article][PubMed]
    [Google Scholar]
  36. Sarwar A, Katas H, Zin NM. Antibacterial effects of chitosan–tripolyphosphate nanoparticles: impact of particle size molecular weight. J Nanopart Res 2014; 16:2517 [View Article]
    [Google Scholar]
  37. Anitha A, Divya Rani VV, Krishna R, Sreeja V, Selvamurugan N et al. Synthesis, characterization, cytotoxicity and antibacterial studies of chitosan, O-carboxymethyl and N,O-carboxymethyl chitosan nanoparticles. Carbohydr Polym 2009; 78:672–677 [View Article]
    [Google Scholar]
  38. Ravikumara NR, Madhusudhan B. Chitosan nanoparticles for tamoxifen delivery and cytotoxicity to MCF-7 and Vero cells. Pure Appl Chem 2011; 83:2027–2040 [View Article]
    [Google Scholar]
  39. Qi L, Xu Z, Jiang X, Li Y, Wang M. Cytotoxic activities of chitosan nanoparticles and copper-loaded nanoparticles. Bioorg Med Chem Lett 2005; 15:1397–1399 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.000445
Loading
/content/journal/jmm/10.1099/jmm.0.000445
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error