EVALUATION OF ANTIBACTERIAL ACTIVITY OF SILVER NANOPARTICLES AGAINST METHICILLIN-RESISTANT STAPHYLOCOCCUS AUREUS AND DETECTION OF VIRULENCE FACTORS - NUCLEASE, PHOSPHATASE, AND BIO FILM PRODUCTION

Authors

  • Manipriya B Department of Microbiology, Saveetha Medical College and Hospital, Thandalam, Kanchipuram, Tamil Nadu, India.
  • Tasneem Banu Department of Microbiology, Saveetha Medical College and Hospital, Thandalam, Kanchipuram, Tamil Nadu, India.
  • Prem Kumar L Department of Microbiology, Saveetha Medical College and Hospital, Thandalam, Kanchipuram, Tamil Nadu, India.
  • Kalyani M Department of Microbiology, Saveetha Medical College and Hospital, Thandalam, Kanchipuram, Tamil Nadu, India.

DOI:

https://doi.org/10.22159/ajpcr.2018.v11i5.24097

Keywords:

Nanoparticles, Silver nanoparticles, Methicillin-resistant Staphylococcus aureus, Biofilm, Nuclease, Phosphatase

Abstract

 Objective: To determine the virulence factors-biofilm, nuclease and phosphatase production in Staphylococcus aureus isolates. To determine the effect of silver nano particles and antibiotics on MRSA by MIC determination and kirby baeur method respectively and finally to compare antibacterial activity of silver nano particles and antibiotics.

Methods: In the present study, we explore the antibacterial activity of silver nanoparticles (Ag-NPs) dispersion (10 nm) against reference strain and clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA). We evaluated the antibacterial activity of Ag-NPs against MRSA isolated from patients in Saveetha Medical College and Hospital, a tertiary care centre in Chennai, Tamil Nadu. The bactericidal activity of different concentrations of Ag-NPs (200, 100, 50, 25, 12.5, 6.25, 3.125, and 1.5625 μg/ml) was tested by determining MIC using microbroth dilution and MBC by agar dilution methods.. In addition, the virulence factors phosphatase, nuclease, and biofilm production were tested.

Result: The values of minimal inhibitory concentration and minimal bactericidal concentration of Ag-NPs against all clinical isolates of MRSA and a single of S. aureus were found in the range of 12.5–50 μg/ml and 12.5–25 μg/ml, respectively, indicating very good bactericidal activity. Ag-NPs with the highest concentration showed almost no growth for up to 16 h representing a bactericidal effect at this concentration. Effect was proportional to dose since 50.0 μg/ml was the most effective dose since the bacterial population did not recover and 12.5 μg/ml was the least effective. All the MRSA isolates were positive for the virulence factors.

Conclusion: The study result suggests that Ag-NPs could be used as an effective alternative antibacterial agent.

Downloads

Download data is not yet available.

Author Biography

Manipriya B, Department of Microbiology, Saveetha Medical College and Hospital, Thandalam, Kanchipuram, Tamil Nadu, India.

Assistant professor , Department of Microbiology

References

Cosgrove SE, Sakoulas G, Perencevich EN, Schwaber MJ, Karchmer AW, Carmeli Y, et al. Comparison of mortality associated with methicillin-resistant and methicillin-susceptible Staphylococcus aureus bacteremia: A meta-analysis. Clin Infect Dis 2003;36:53-9.

Grundmann H, Aires-de-Sousa M, Boyce J, Tiemersma E. Emergence and resurgence of meticillin-resistant Staphylococcus aureus as a public-health threat. Lancet 2006;368:874-85.

Wu SW, de Lencastre H, Tomasz A. Recruitment of the mecA gene homologue of Staphylococcus sciuri into a resistance determinant and expression of the resistant phenotype in Staphylococcus aureus. J Bacteriol 2001;183:2417-24.

Ayliffee GA. The progressive intercontinental spread of methicillin-resistant Staphylococcus aureus. Clin Infect Dis 1997;24:S74-9.

Johnson AP, Pearson A, Duckworth G. Surveillance and epidemiology of MRSA bacteraemia in the UK. J Antimicrob Chemother 2005;56:455-62.

Jones N, Ray B, Ranjit KT, Manna AC. Antibacterial activity of znO nanoparticle suspensions on a broad spectrum of microorganisms. FEMS Microbiol Lett 2008;279:71-6.

Kwon AS, Park GC, Ryu SY, Lim DH, Lim DY, Choi CH, et al. Higher biofilm formation in multidrug-resistant clinical isolates of Staphylococcus aureus. Int J Antimicrob Agents 2008;32:68-72.

Lewis K, Klibanov AM. Surpassing nature: Rational design of sterile-surface materials. Trends Biotechnol 2005;23:343-8.

Lewis K. Multidrug tolerance of biofilms and persister cells. Curr Top Microbiol Immunol 2008;322:107-31.

Martino PD, Fursy R, Bret L, Sundararaju B, Phillips RS. Indole can act as an extracellular signal to regulate biofilm formation of escherichia coli and other indole-producing bacteria. Can J Microbiol 2003;49:443-9.

Patel CN, Wortham BW, Lines JL, Fetherston JD, Perry RD, Oliveira MA, et al. Polyamines are essential for the formation of plague biofilm. J Bacteriol 2006;188:2355-63.

McGinnis MW, Parker ZM, Walter NE, Rutkovsky AC, Cartaya-Marin C, Karatan E, et al. Spermidine regulates vibrio cholerae biofilm formation via transport and signaling pathways. FEMS Microbiol Lett 2009;299:166-74.

Kostakioti M, Hadjifrangiskou M, Hultgren SJ. Bacterial biofilms: Development, dispersal, and therapeutic strategies in the dawn of the postantibiotic era. Cold Spring Harb Perspect Med 2013;3:a010306.

Lowy FD. Staphylococcus aureus infections. N Engl J Med 1998;339:520-32.

Ammendolia MG, Di Rosa R, Montanaro L, Arciola CR, Baldassarri L. Slime production and expression of the slime-associated antigen by staphylococcal clinical isolates. J Clin Microbiol 1999;37:3235-8.

Cramton SE, Gerke C, Schnell NF, Nichols WW, Götz F. The intercellular adhesion (ica) locus is present in Staphylococcus aureus and is required for biofilm formation. Infect Immun 1999;67:5427-33.

Arciola CR, Baldassarri L, Montanaro L. Presence of icaA and ica DGenes and slime production in a collection of staphylococcal strains from catheter-associated infections. J Clin Microbiol 2001;39:2151-6.

O’Neill E, Pozzi C, Houston P, Smyth D, Humphreys H, Robinson DA, et al. Association between methicillin susceptibility and biofilm regulation in Staphylococcus aureus isolates from device-related infections. J Clin Microbiol 2007;45:1379-88.

Merz WG, Hay RJ. Topley and Wilson’s Bacteriology. 10th ed. New York: ASM Press; 2005.

Schulman BM, Redfield C, Peng Z, Dobson CM, Kim PS. Different subdomains are most protected from hydrogen exchange in the molten globule and native states of human α-lactalbumin. J Mol Biol 1995;253:651-7.

Collee JG, Fraser AG, Marmion BP, Simmons A. Mackie and McCartney. 14th ed. New Delhi: Churchill Livingstone; 1996.

Thill A, Zeyons O, Spalla O, Chauvat F, Rose J, Auffan M, et al. Cytotoxicity of ceO2 nanoparticles for Escherichia coli. Physico-chemical insight of the cytotoxicity mechanism. Environ Sci Technol 2006;40:6151-6.

Sondi I, Salopek-Sondi B. Silver nanoparticles as antimicrobial agent: A case study on E. coli as a model for gram-negative bacteria. J Colloid Interface Sci 2004;275:177-82.

Lok CN, Ho CM, Chen R, He QY, Yu WY, Sun H, et al. Silver nanoparticles: Partial oxidation and antibacterial activities. J Biol Inorg Chem 2007;12:527-34.

Rangari A, Sharma NK, Ms RG, Thakur R, Singh P. Evaluation and efficacy of in-vitro antibacterial activity of silver nano particles against multidrug resistant bacterial Isolates from skin infections of patients at a tertiary care hospital in western Uttar Pradesh of India. Int J Curr Microbiol Appl Sci 2015;4:764-73.

Morones JR, Elechiguerra JL, Camacho A, Holt K, Kouri JB, Ram´ırez JT, et al. The bactericidal effect of silver nanoparticles. Nanotechnology 2005;16:2346-53.

Kvitek L, PanáÄek A, Soukupová J, Kolář M, VeÄeÅ™ová R, Prucek R, et al. Effect of surfactants and polymers on stability and antibacterial activity of silver nanoparticles (NPs). J Phys Chem 2008;112:5825-34.

Nanda A, Saravanan M. Biosynthesis of silver nanoparticles from Staphylococcus aureus and its antimicrobial activity against MRSA and MRSE. Nanomedicine 2009;5:452-6.

Panacek A, Kvítek L, Prucek R, Kolar M, Vecerova R, Pizúrova N, et al. Silver colloid nanoparticles: Synthesis, characterization, and their antibacterial activity. J Phys Chem B 2006;110:16248-53.

Silver S. Bacterial silver resistance: Molecular biology and uses and misuses of silver compounds. FEMS Microbiol Rev 2003;27:341-53.

Choi O, Deng KK, Kim NJ, Ross L Jr., Surampalli RY, Hu Z, et al. The inhibitory effects of silver nanoparticles, silver ions, and silver chloride colloids on microbial growth. Water Res 2008;42:3066-74.

Tamayo LA, Zapata PA, Vejar ND, Azócar MI, Gulppi MA, Zhou X, et al. Release of silver and copper nanoparticles from polyethylene nanocomposites and their penetration into listeria monocytogenes. Mater Sci Eng C Mater Biol Appl 2014;40:24-31.

Tian N, Zhou ZY, Sun SG, Ding Y, Wang ZL. Synthesis of tetrahexahedral platinum nanocrystals with high-index facets and high electro-oxidation activity. Science 2007;316:732-5.

French GL. Bactericidal agents in the treatment of MRSA infections – The potential role of daptomycin. J Antimicrob Chemother 2006;58:1107-17.

Gong P, Li H, He X, Wang K, Hu J, Tan W, et al. Preparation and antibacterial activity of Fe3O4@Ag nanoparticles. Nanotechnology 2007;18:1-17.

Christensen GD, Simpson WA, Younger JJ, Baddour LM, Barrett FF, Melton DM, et al. Adherence of cogulase negative staphylococi to plastic tissue cultures: A quantitative model for the adherence of staphylococci to medical devices. J Clin Microbiol 1985;22:996-1006.

Stepanovic S, Vuković D, Hola V, Di Bonaventura G, Djukić S, Cirković I, et al.APMIS Quantification of biofilm in microtiter plates: Overview of testing conditions and practical recommendations for assessment of biofilm APMIS 2007;115:891-9.

Mathur T, Singhal S, Khan S, Upadhyay DJ, Fatma T, Rattan A, et al. Detection of biofilm formation among the clinical isolates of staphylococci: An evaluation of three different screening methods. Indian J Med Microbiol 2006;24:25-9.

Grinholc M, Wegrzyn G, Kurlenda J. Evaluation of biofilm production and prevalence of the icaD gene in methicillin-resistant and methicillin-susceptible Staphylococcus aureus strains isolated from patients with nosocomial infections and carriers. FEMS Immunol Med Microbiol 2007;50:375-9.

Ansari MA, Khan HM, Khan AA, Malik A, Sultan A, Shahid M, et al. Evaluation of antibacterial activity of silver nanoparticles against MSSA and MSRA on isolates from skin infections. Biol Med 2011;3:141-6.

Li WR, Xie XB, Shi QS, Duan SS, Ouyang YS, Chen YB, et al. Antibacterial effect of silver nanoparticles on Staphylococcus aureus. Biometals 2011;24:135-41.

Kim JS, Kuk E, Yu KN, Kim JH, Park SJ, Lee HJ, et al. Antimicrobial effects of silver nanoparticles. Nanomedicine 2007;3:95-101.

Ahmad MI, Srikanth. Prevalence and antimicrobial susceptibility of methicillin resistant Staphylococcus aureus and coagulase negative staphylococci in tertiary care hospital. Asian J Pharm Clin Res 2013;6 Suppl 3:231-4.

Mary RN, Banu N. Screening of anti biofilm and anti-quorom sensing potential of vitex trifolia in Psuedomonas aeruginosa. Int J Pharm Pharm Sci 2005;7:242-5.

Turkyilmaz S, Kaya O. Determination of some virulence factors in Staphylococcus Spp. isolated from various clinical samples. Turk J Vet Anim Sci 2006;30:127-32.

Agarwal H, Gayathri M. Biological synthesis of nanoparticles from medicinal plants and its uses in inhibiting biofilm formation. Asian J Pharm Clin Res 2017;10:64-8.

Sunita P, Palaniswamy M. Size dependent application of biologically synthesized silver nanoparticles against bacterial skin pathogens. Asian J Pharm Clin Res 2017;10:192-5.

Published

01-05-2018

How to Cite

B, M., T. Banu, P. Kumar L, and K. M. “EVALUATION OF ANTIBACTERIAL ACTIVITY OF SILVER NANOPARTICLES AGAINST METHICILLIN-RESISTANT STAPHYLOCOCCUS AUREUS AND DETECTION OF VIRULENCE FACTORS - NUCLEASE, PHOSPHATASE, AND BIO FILM PRODUCTION”. Asian Journal of Pharmaceutical and Clinical Research, vol. 11, no. 5, May 2018, pp. 224-9, doi:10.22159/ajpcr.2018.v11i5.24097.

Issue

Section

Original Article(s)