ISSN: 2455-5363
Global Journal of Infectious Diseases and Clinical Research
Review Article       Open Access      Peer-Reviewed

A Short Review on Advances in Nanosystems Emerging as an Effective Approaches to Control Pathogenesis of Staphylococcus spp

Ved Prakash Giri1,3, Pallavi Shukla1,2, Ashutosh Tripathi1,2, Navinit Kumar1,2, Priya Verma1,2, Shipra Pandey1,2 and Aradhana Mishra1,2*

1Principal Scientist, Division of Microbial Technology, CSIR- National Botanical Research Institute, Lucknow, 226001, India
2Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
3Department of Botany, Lucknow University, Hasanganj, Lucknow, 226007, India
*Corresponding author: Dr. Aradhana Mishra, Principal Scientist, Division of Microbial Technology, CSIR- National Botanical Research Institute, Lucknow, 226001, India, Tel: 91 522 2297987; Fax: +91 522 2205839; Email: mishra.a@nbri.res.in, mishramyco@yahoo.com
Received: 07 July, 2021 | Accepted: 17 August, 2021 | Published: 19 August, 2021
Keywords: Nanosystems; Staphylococcus spp; Pathogenesis mechanisms; Nanomaterials

Cite this as

Giri VP, Shukla P, Tripathi A, Kumar N, Mishra A, et al. (2021) A Short Review on Advances in Nanosystems Emerging as an Effective Approaches to Control Pathogenesis of Staphylococcus spp. Glob J Infect Dis Clin Res 7(1): 049-055. DOI: 10.17352/2455-5363.000046

Nanomaterials in drug delivery systems are emerging as a potential source of treatments. The bioavailability of nano-carriers depends upon various factors including their mode of administration, target specificity, availability of active compound and compatibility with biological system. Due to ingenious survival strategies many antibiotics become ineffective in the treatment of Staphylococcus spp. causing diseases. Though, nanosystems have been developed as an emerging approach to vanquish the obstacles for the treatment of Staphylococcus spp infections. Due to their encouraging and substantial outcomes, this review focused on the exploration of advances in nanosystems and their curing mechanism to control the infection of Staphylococcus spp. Several nanosystems are previously reported and compiled herein, which may use as a treatment of Staphylococcus causing diseases as well as inhibition of biofilm formation. Pathogenesis mechanisms such as immune clearance, membrane permeability and production of β-lactamase also described briefly in this review.

Introduction

The Staphylococcus is a gram positive facultative aerobic bacteria. It belongs to the family Staphylococcaceae and order Bacillales. It mainly reside on the skin and mucous membrane of humans and other animal. Staphylococcus is an opportunistic pathogen mainly colonize on human anterior nares and causes life threatening bloodstream infection like sepsis and endocarditis. It causes hospital-acquired infections mainly in soft tissue by confiscating the host defense and coagulation systems [1]. The infections creates high mortality rate despite of the proper treatment. Staphylococcus aureus is one of the most fatal bacteria, approx 20% to 50% mortality has estimated [2]. It causes inflammation and immunosuppression which leads to the disseminated intravascular coagulation (DIC), damage the endothelium layer and blocks blood stream as well as resulted as oxygen depletion in organs [3,4]. The adhesion process is the key step for the pathogenesis of S. aureus by secreting many toxins. They also secrete many factors like fibronectin binding protein A (Fnbp A), Fnbp B and teichoic acid which promote colonization at infection site and resist to immune response of the host [5]. The S. aureus develops resistance to β-lactams, daptomycin and glycopeptide antibiotics. Moreover, Methicillin-Resistant Staphylococcus aureus (MRSA) also develops an antibiotic resistance by over secreation of β- lactamases and reducing the efficacy of antibiotics.

Due to ingenious survival strategies many antibiotics are ineffective in the treatment of S. aureus. Therefore, deal with nanomaterials have developed as an emerging approach to vanquish the obstacles for the treatment of S. aureus infections. Nanosystems have ability to inhibit the biofilm formation, cell wall penetration, enhanced intracellular retention and improve antibacterial activity of the loaded antimicrobial agents [6]. However, several nanoparticles passively accumulates in targeted organs because of their own characteristics, such as nanosize, surface charge, and large surface area. While, modified nanomaterials could increase the membrane transportation by actively identify the receptors of host cells as well as bacterial cells. Currently, several microbicidal agents are conjugated with nanocarriers to increase the pharmacological activities against Staphylococcus spp. Therefore, nanosystems drug delivery advocated as an ideal weapon to control the pathogenicity of Staphylococcus spp.

Nanomaterials as chemical substances or biomaterials that are manufactured and used at a very small scale. Nanomaterials are develop to exhibit novel characteristics such as increased strength, chemical reactivity or conductivity as compared to the same material without nanoscale features. Due to their large surface area and easy surface modification, nanoparticles also used as nanocarriers to carry the drugs to targeted site in drug delivery system. Several metallic, magnetic, silica and carbon based nanoparticles have been designed for drug delivery applications [7,8].

This review describe the pathogenesis mechanisms of Staphylococcus such as immune clearance, membrane permeability and production of β-lactamase. We have compiled the several nanosystems which are previously known as a bactericidal agent for Staphylococcus spp. and their action mechanism also be summarized herein.

Pathogenesis mechanisms of Staphylococcus which causes infection

By immune clearance: In the bloodstream, Kupffer cells efficiently regulates Staphylococcus survivability by phagocytosis while sometimes the phagocytosis process has compromises then staphylococci certainly survive and multiply inside the cells. Thus, they release from intracellular niche and encounters by the peritoneal macrophages and bloodstream neutrophils [9]. An essential attributes of Staphylococcus requires to create infection and coordinate with timely expression of virulence factors along with relevant genes. Several reports are highlighted the genetic adaptation system ‘accessory gene regulator’ (Agr) or quorum-sensing system in S. aureus which requires for secretion of number of toxins and other soluble virulence factors [10]. It senses an auto inducing peptide (AIP) secreted by the S. aureus itself. The activation of Agr signaling occurs when AIP reaches a critical concentration due to high bacterial density. An active Agr system is essential for intracellular survival of S. aureus or inside the phagocytes cells [11].

By membrane permeability: The cell membrane permeability is another factor which directly relates to the bacterial survivability. Drug absorption is reduces when the energy metabolism of the bacteria is affected and creates a lower cell membrane permeability and leads to the drug resistance [12].

Long term exposure of drugs activates the efflux system which enhance the efflux of drugs and develop a drug resistance in the bacteria [13]. The efflux proteins exchanges takes place by electrochemical gradient formed by H+ on both side of cell membrane [14]. Majorly, three multidrug-pumping proteins viz; QacA, NorA, and Smr present on the Staphylococcus aureus cell membrane (Foster, 2016). Among these, QacA is an important factor in MRSA [15].

By production of beta lactamase: The β-lactamase causes hydrolysis of β-lactam antibiotics and develops a lethal effect on bacteria by two mechanism (i) by binding to the penicillin binding protein which repress mucin synthesis in cell wall and leads to the cell wall disruption as well as bacterial lysis (ii) by trigger autolytic enzyme activity of bacteria [16]. Moreover, MRSA also develops an antibiotic resistance by over secreation of β- lactamases. It reduces the effect of antibiotics by hydrolysis of β-lactamase and resulted as inactivation of antibiotics. β-lactamase binds to the antibiotics and prevents them to reach the target site .

Action mechanism of nanomaterials to prevention of Staphylococcus spp. infection

Nano pharmaceuticals could efficiently administer to the human body by orally, intravenously, respiratory system and topically [17]. The size, shape, surface charge, chemical composition and hydrophobicity of nanosystems influences to the intestinal mucus. The mechanism of action is different with different set of nanomaterials for example liposome nanomaterials with PEG coating are easily able to penetrate in mucus layer than nanomaterials coated with chitosan. The negative charges presents on the surface of nanomaterial as well as mucus that causes repulsion and hydrophilicity of the mucus surface [18]. A previous study suggested that zinc oxide nanoparticles doped with digestive enzyme ‘pancreatin’ which shows potential antibacterial activity against Methicillin Resistant Staphylococcus aureus (MRSA). It induces the oxidative stress, membrane potential alteration and membrane damages. Their synergistic effects resulted as degradation of biofilm of pathogens through protease and amylase lipase activity of ‘pancreatin’. The reduction of membrane potential leads to ROS generation and alters the homeostasis of the pathogen, oxidization of lipid content and resulted as disintegration of bacterial cell wall [19].

Metal nanoparticles

The metal nanoparticles exhibits potential antibacterial activity and helps to overcome resistance in pathogens. Their mechanism of action involves in metal ion release, cell wall and membrane disintegration, ROS generation, intracellular penetration and DNA damages [20]. Silver nanoparticles are widely accepted for their effective antibacterial activity by altering the cell wall protein, inhibits cell division, disrupt signal transduction and cause ROS production [21]. Recently our research team also reported the strong bactericidal activity of biogenic silver nanoparticles than their chemical counterparts [22] Figure 1.

The gold nanoparticles (AuNPs) shows promising results due to their high biocompatibility property [23]. Ultrasound-triggered nanosystem also used to treat MRSA infection by modifying polyethylenimine grafted bismuth oxybromide (BiOBr) nanoplates with

Fe3. [24]. Likewise, Zinc-doped Prussian Blue (ZnPB) had a great photothermal anti-MRSA activity.

Carbon based nanomaterial

Carbon Based Nanomaterial (CBN) including Graphene Oxide (GO), Reduced Graphene Oxide (RGO), Graphene Quantum Dots (GQDs), Carbon Quantum Dots (CQDs), Carbon Nanotubes (CNTs), and Carbon Nanofibers (CNFs) are also used as potent antimicrobial agent. Among these, GO have excellent chemical and mechanical properties and it can penetrate the cell membranes as well as leading cellular damages of the bacteria [25]. The CNB have high permeability which makes them efficient to penetrate the thick biofilm. At present, polymer-based nanocomposites also used as antibiotic carriers to treat intracellular MRSA. The diblock guanidinium polymer has been found efficient internalization in keratinocytes and inhibits the growth of intracellular MRSA [25]. Several other nanosystems are also have been reported for prevention and treatment of Staphylococcus spp. causing diseases listed in Table 1.

Advantages and disadvantages of nanosystems

Increase in antibiotic resistance in bacteria emerges as a greatest challenge to human health. Though, deal with nanomedicine plays a vital role in increasing the potency of existing therapies, by increasing stability and physiochemical properties of antibiotics, by biofilm internalization, prolongation of antibiotic release and targeted delivery of drug to the site of infection [58]. Size and surface potential of nanomaterials drives interaction with various component of the tissue and regulates intracellular uptake and biodistribution [59]. Hydrophobic property of nanomaterials helps in targeting the drugs to the targeted site and enhance the adhesion efficacy [58]. Nano-sized systems have ability to impede the instigation of resistance as well as vanquishing the resistance strategies of bacteria [60].

Despite of significant improvements in antibiotics delivery and stability, nanosystems also suffering from many drawbacks that limiting their efficient usage. Physical and chemical instability of nanoparticles, antibiotic leakage from liposomes due to incompatible physiological conditions and low loading capacity [62-72] are listed as major demerits of nanosystems. Several lipids sensitivity occurs due to high temperatures as well as fabrication techniques are very complex, expensive, and difficult to be scaled up [63].

Conclusion

The strategies highlighted in this review based on using different nanosystems to endure antibiotic resistance against Staphylococcus spp. several antibiotics and bioactive materials showed enhanced bactericidal activity when they were loaded with nanosystems. Zeta potential, particle size, and solubility are the important physico-chemical factors for drug delivery. This review will helps the readers to explore the different nanosystems which have preventive approaches to control the Staphylococcus spp. infection. Majorly, immune clearance, membrane permeability and production of β-lactamase employed in Staphylococcus pathogenicity. Description of pathogenesis mechanism of Staphylococcus and action mechanism of nanomaterial could provide a concept between interaction of nanosystems and pathogen in human body.

  1. Dryden MS (2009) Skin and soft tissue infection: microbiology and epidemiology. Int J Antimicrob Agents 34: S2-S7. Link: https://bit.ly/2W4jk5V
  2. Asgeirsson H, Thalme A, Weiland O (2018) Staphylococcus aureus bacteraemia and endocarditis–epidemiology and outcome: a review. Infect Dis 50: 175-192. Link: https://bit.ly/3iYp9Le
  3. Angus DC, Van der Poll T (2013) Severe sepsis and septic shock. N Engl J Med 369: 840-851. Link: https://bit.ly/2W6NiWW
  4. Löffler B, Tuchscherr L, Niemann S, Peters G (2014) Staphylococcus aureus persistence in non-professional phagocytes. Int J Med Microbiol 304: 170-176.
  5. Zhou K, Li C, Chen D, Pan Y, Tao Y, et al. (2018) A review on nanosystems as an effective approach against infections of Staphylococcus aureus. Int J Nanomedicine 13: 7333.
  6. Park SB, Steadman CS, Chaudhari AA, Pillai SR, Singh SR, et al. (2018) Proteomic analysis of antimicrobial effects of pegylated silver coated carbon nanotubes in Salmonella enterica serovar Typhimurium. Journal of Nanobiotechnology 16: 1-14. Link: https://bit.ly/3z4nLMH
  7. Kumar R, Dalvi SV, Siril PF (2020) Nanoparticle-based drugs and formulations: current status and emerging applications. ACS Applied Nano Materials 3: 49444961. Link: https://bit.ly/2W5ciOu
  8. Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, et al. (2021) Nanocarrier vaccines for SARSCoV-2. Adv Drug Deliv Rev 171: 215-239. Link: https://bit.ly/3gfpd7L
  9. Jorch SK, Surewaard BG, Hossain M, Peiseler M, Deppermann C, et al. (2019) Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination. J Clin Invest 129: 4643-4656. Link: https://bit.ly/3swN9Ze
  10. Jenul C, Horswill AR (2019) Regulation of Staphylococcus aureus virulence. Microbiol Spectr 7: 10. Link: https://bit.ly/3AODAHG
  11. Painter KL, Krishna A, Wigneshweraraj S, Edwards AM (2014) What role does the quorum-sensing accessory gene regulator system play during Staphylococcus aureus bacteremia?. Trends Microbiol 22: 676-685. Link: https://bit.ly/2XG0ini
  12. Anuj SA, Gajera HP, Hirpara DG, Golakiya BA (2019) Interruption in membrane permeability of drug-resistant Staphylococcus aureus with cationic particles of nano-silver. Eur J Pharm Sci 127: 208-216. Link: https://bit.ly/3AZzYTD
  13. Zárate SG, Morales P, Świderek K, Bolanos-Garcia VM, Bastida A (2019) A molecular modeling approach to identify novel inhibitors of the major facilitator superfamily of efflux pump transporters. Antibiotics 8: 25. Link: https://bit.ly/3CYswd8
  14. Matano LM, Morris HG, Hesser AR, Martin SE, Lee W, et al. (2017) Antibiotic that inhibits the ATPase activity of an ATP-binding cassette transporter by binding to a remote extracellular site. J Am Chem Soc 139: 10597-10600. Link: https://bit.ly/3iZzu9S
  15. Nakaminami H, Takadama S, Okita M, Sasaki M, Noguchi N (2019) Fast-acting bactericidal activity of olanexidine gluconate against qacA/B-positive methicillin-resistant Staphylococcus aureus. Journal of medical microbiology 68: 957-960.
  16. Matono T, Nagashima M, Mezaki K, Motohashi A, Kutsuna S, et al. (2018) Molecular epidemiology of β-lactamase production in penicillin-susceptible Staphylococcus aureus under high-susceptibility conditions. J Infect Chemother 24: 153-155. Link: https://bit.ly/3xZL0Gq
  17. Staroń A, Długosz O, Pulit-Prociak J, Banach M (2020) Analysis of the Exposure of Organisms to the Action of Nanomaterials. Materials 13: 349. Link: https://bit.ly/3ASRwR5
  18. Falavigna M, Klitgaard M, Brase C, Ternullo S, Škalko-Basnet N, et al. (2018) Mucus-PVPA (mucus phospholipid vesicle-based permeation assay): an artificial permeability tool for drug screening and formulation development. International journal of pharmaceutics 537: 213-222. Link: https://bit.ly/3CYssdo
  19. Banerjee S, Vishakha K, Das S, Dutta M, Mukherjee D, et al. (2020) Antibacterial, anti-biofilm activity and mechanism of action of pancreatin doped zinc oxide nanoparticles against methicillin resistant Staphylococcus aureus. Colloids and Surfaces B: Biointerfaces 190: 110921. Link: https://bit.ly/3j0AL0e
  20. Fanoro OT, Oluwafemi OS (2020) Bactericidal antibacterial mechanism of plant synthesized silver, gold and bimetallic nanoparticles. Pharmaceutics 12: 1044. Link: https://bit.ly/2W3IKk8
  21. Rahimi M, Noruzi EB, Sheykhsaran E, Ebadi B, Kariminezhad Z, et al. (2020) Carbohydrate polymer-based silver nanocomposites: Recent progress in the antimicrobial wound dressings. Carbohydr Polym 231: 115696. Link: https://bit.ly/3z2G4lk
  22. Kumari M, Shukla S, Pandey S, Giri VP, Bhatia A, et al. (2017) Enhanced cellular internalization: a bactericidal mechanism more relative to biogenic nanoparticles than chemical counterparts. ACS Appl. Mater. Interfaces 9: 4519-4533.
  23. Okoampah E, Mao Y, Yang S, Sun S, Zhou C (2020) Gold nanoparticlesbiomembrane interactions: from fundamental to simulation. Colloids and Surfaces B: Biointerfaces 111312. Link: https://bit.ly/2XFyOyb
  24. Song M, Cheng Y, Tian Y, Chu C, Zhang C, et al. (2020) Sonoactivated Chemodynamic Therapy: A Robust ROS Generation Nanotheranostic Eradicates MultidrugResistant Bacterial Infection. Advanced Functional Materials 30: 2003587. Link: https://bit.ly/3k6Tfvl
  25. Gao Y, Chen Y, Cao Y, Mo A, Peng Q (2020) Potentials of nanotechnology in treatment of methicillin-resistant Staphylococcus aureus. Eur J Med Chem 213: 113056. Link: https://bit.ly/3mgQU3Q
  26. He Q, Zhang L, Song L, Zhang X, Liu D, et al. (2021) Inactivation of Staphylococcus aureus using ultrasound in combination with thyme essential oil nanoemulsions and its synergistic mechanism. LWT 147: 111574. Link: https://bit.ly/3z13LKW
  27. Guan G, Zhang L, Zhu J, Wu H, Li W, et al. (2021) Antibacterial properties and mechanism of biopolymer-based films functionalized by CuO/ZnO nanoparticles against Escherichia coli and Staphylococcus aureus. Journal of Hazardous Materials 402: 123542. Link: https://bit.ly/3gff22Y
  28. Sivakumar M, Surendar S, Jayakumar M, Seedevi P, Sivasankar P, et al. (2021) Parthenium hysterophorus mediated synthesis of silver nanoparticles and its evaluation of antibacterial and antineoplastic activity to combat liver cancer cells. Journal of Cluster Science 32: 167-177. Link: https://bit.ly/3ssCQVX
  29. Navarro-López DE, Garcia-Varela R, Ceballos-Sanchez O, Sanchez-Martinez A, Sanchez-Ante G, et al. (2021) Effective antimicrobial activity of ZnO and Yb-doped ZnO nanoparticles against Staphylococcus aureus and Escherichia coli. Materials Science and Engineering: C 123: 112004. Link: https://bit.ly/3z2qG8z
  30. Ma S, Moser D, Han F, Leonhard M, Schneider-Stickler B, et al. (2020) Preparation and antibiofilm studies of curcumin loaded chitosan nanoparticles against polymicrobial biofilms of Candida albicans and Staphylococcus aureus. Carbohydrate Polymers 241: 116254. Link: https://bit.ly/3mgQWZw
  31. Shabangu SM, Babu B, Soy RC, Oyim J, Amuhaya E, et al. (2020) Susceptibility of Staphylococcus aureus to porphyrin-silver nanoparticle mediated photodynamic antimicrobial chemotherapy. Journal of Luminescence 222: 117158. Link: https://bit.ly/3k5tCLl
  32. Fulaz S, Devlin H, Vitale S, Quinn L, O’Gara JP, et al. (2020) Tailoring nanoparticle-biofilm interactions to increase the efficacy of antimicrobial agents against Staphylococcus aureus. Int J Nanomedicine 15: 4779. Link: https://bit.ly/3ARvZIA
  33. Rajkumar PV, Prakasam A, Rajeshkumar S, Gomathi M, Anbarasan PM, et al. (2020) Green synthesis of silver nanoparticles using Gymnema sylvestre leaf extract and evaluation of its antibacterial activity. South African Journal of Chemical Engineering 32: 1-4. Link: https://bit.ly/2W8888D
  34. Akter S, Huq MA (2020) Biologically rapid synthesis of silver nanoparticles by Sphingobium sp. MAH-11T and their antibacterial activity and mechanisms investigation against drug-resistant pathogenic microbes. Artif Cells Nanomed Biotechnol 48: 672-682. Link: https://bit.ly/3z0cQ6x
  35. Khan N, Kumar D, Kumar P (2020) Silver nanoparticles embedded guar gum/gelatin nanocomposite: Green synthesis, characterization and antibacterial activity. Colloid and Interface Science Communications 35: 100242. Link: https://bit.ly/2W7FTaf
  36. Nirmala MJ, Durai L, Gopakumar V, Nagarajan R (2020) Preparation of Celery Essential Oil-Based Nanoemulsion by Ultrasonication and Evaluation of Its Potential Anticancer and Antibacterial Activity. International Journal of Nanomedicine 15: 7651. Link: https://bit.ly/3ARwa6I
  37. Li CH, Landis RF, Makabenta JM, Nabawy A, Tronchet T, Archambault D, et al. (2021) Nanotherapeutics using all-natural materials. Effective treatment of wound biofilm infections using crosslinked nanoemulsions. Materials Horizons. Link: https://rsc.li/2XJ0TVr
  38. Li J, Zhong W, Zhang K, Wang D, Hu J, et al. (2020) BiguanideDerived Polymeric Nanoparticles Kill MRSA Biofilm and Suppress Infection In Vivo. ACS Appl Mater Interfaces 12: 21231-21241. Link: https://bit.ly/3z0cZH7
  39. Hamida RS, Ali MA, Goda DA, Khalil MI, Al-Zaban MI (2020) Novel biogenic silver nanoparticle-induced reactive oxygen species inhibit the biofilm formation and virulence activities of methicillin-resistant Staphylococcus aureus (MRSA) strain. Front Bioeng Biotechnol 8: 433. Link: https://bit.ly/3AVxVQo
  40. Iqbal H, Khan BA, Khan ZU, Razzaq A, Khan NU, et al. (2020) Fabrication, physical characterizations and in vitro antibacterial activity of cefadroxil-loaded chitosan/poly (vinyl alcohol) nanofibers against Staphylococcus aureus clinical isolates. International Journal Biological Macromolecules 144: 921-931. Link: https://bit.ly/3kb1fM5
  41. Hassan D, Omolo CA, Fasiku VO, Mocktar C, Govender T (2020) Novel chitosan-based pH-responsive lipid-polymer hybrid nanovesicles (OLA-LPHVs) for delivery of vancomycin against methicillin-resistant Staphylococcus aureus infections. International Journal Biological Macromolecules 147: 385-398. Link: https://bit.ly/2W4jZnV
  42. Oves M, Rauf M, Ansari MO, Aslam Parwaz Khan AA, Qari H, et al. (2020) Graphene Decorated Zinc Oxide and Curcumin to Disinfect the Methicillin-Resistant Staphylococcus aureus. Nanomaterials 10: 1004. Link: https://bit.ly/3xZB2Vx
  43. Slavin YN, Ivanova K, Hoyo J, Perelshtein I, Owen G, et al. (2021) Novel Lignin-Capped Silver Nanoparticles against Multidrug-Resistant Bacteria. ACS Appl Mater Interfaces 13: 22098-22109. Link: https://bit.ly/3j1AiLr
  44. Omran Z, Bader A, Porta A, Vandamme T, Anton N, et al. (2020) Evaluation of Antimicrobial Activity of Triphala Constituents and Nanoformulation. Evidence-Based Complementary and Alternative Medicine 2020: 6976973. Link: https://bit.ly/3AWJO8O
  45. Mazur P, Skiba-Kurek I, Mrowiec P, Karczewska E, Drożdż R (2020) Synergistic ROS-associated antimicrobial activity of silver nanoparticles and gentamicin against Staphylococcus epidermidis. Int J Nanomedicine 15: 3551-3562. Link: https://bit.ly/3z2GtUS
  46. Swolana D, Kępa M, Idzik D, Dziedzic A, Kabała-Dzik A, et al. (2020) The antibacterial effect of silver nanoparticles on Staphylococcus epidermidis strains with different biofilm-forming ability. Nanomaterials 10: 1010. Link: https://bit.ly/3ssUrgx
  47. Arshad H, Sami MA, Sadaf S, Hassan U (2021) Salvadora persica mediated synthesis of silver nanoparticles and their antimicrobial efficacy. Scientific Reports 11: 1-11. Link: https://go.nature.com/2WbE1Ny
  48. Marangon CA, Martins VC, Ling MH, Melo CC, Plepis AMG, et al. (2020) Combination of rhamnolipid and chitosan in nanoparticles boosts their antimicrobial efficacy. ACS Appl Mater Interfaces 12: 5488-5499.
  49. Boomi P, Ganesan R, Poorani GP, Jegatheeswaran S, Balakumar C, et al. (2020) Phyto-Engineered Gold Nanoparticles (AuNPs) with potential antibacterial, antioxidant, and wound healing activities under in vitro and in vivo Conditions. Int J Nanomedicine 15: 7553.
  50. Amin RM, Mohamed MB, Ramadan MA, Verwanger T, Krammer B (2009) Rapid and sensitive microplate assay for screening the effect of silver and gold nanoparticles on bacteria. Nanomedicine 4: 637-643. Link: https://bit.ly/3AS7UBo
  51. Gan L, Geng A, Jin L, Zhong Q, Wang L, et al. (2020) Antibacterial nanocomposite based on carbon nanotubes–silver nanoparticles-co-doped polylactic acid. Polymer Bulletin 77: 793-804. Link: https://bit.ly/3suUJmO
  52. Maccelli A, Vitanza L, Imbriano A, Fraschetti C, Filippi A, et al. (2020) Satureja montana L. essential oils: chemical profiles/phytochemical screening, antimicrobial activity and O/W NanoEmulsion formulations. Pharmaceutics 12: 7. Link: https://bit.ly/2Xv8qa0
  53. Sumitha S, Vasanthi S, Shalini S, Chinni SV, Gopinath SCB, et al. (2019) Durio zibethinus rind extract mediated green synthesis of silver nanoparticles: Characterization and biomedical applications. Pharmacognosy Magazine 15: 52. Link: https://bit.ly/3iWPdX9
  54. Rashid S, Azeem M, Khan SA, Shah MM, Ahmad R (2019) Characterization and synergistic antibacterial potential of green synthesized silver nanoparticles using aqueous root extracts of important medicinal plants of Pakistan. Colloids and Surfaces B: Biointerfaces 179: 317-325. Link: https://bit.ly/3mgl5rC
  55. Darroudi M, KhandaKhandan Nasab N, Salimizand H, Dehnad A (2019) Green synthesis and antibacterial activity of zinc selenide (ZnSe) nanoparticles. Nanomedicine Journal 6: 258-262. Link: https://bit.ly/3CZwkut
  56. Orellano MS, Isaac P, Breser ML, Bohl LP, Conesa A, et al. (2019) Chitosan nanoparticles enhance the antibacterial activity of the native polymer against bovine mastitis pathogens. Carbohydrate polymers 213: 1-9. Link: https://bit.ly/3xZLyfs
  57. Wang J, Wang Y, Zhang D, Chen C (2020) Intrinsic oxidase-like nanoenzyme Co4S3/Co (OH) 2 hybrid nanotubes with broad-spectrum antibacterial activity. ACS Appl Mater Interfaces 12: 29614-29624. Link: https://bit.ly/3AUPrEs
  58. Patra JK, Das G, Fraceto LF, Campos EVR, del Pilar Rodriguez-Torres M, et al. (2018) Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology 16: 71. Link: https://bit.ly/37T8vGn
  59. Dacoba TG, Olivera A, Torres D, Crecente-Campo J, Alonso MJ (2017) Modulating the immune system through nanotechnology. Semin Immunol 34: 78-102. Link: https://bit.ly/3xZuqGA
  60. Baptista PV, McCusker MP, Carvalho A, Ferreira DA, Mohan NM, et al. (2018) Nano-strategies to fight multidrug resistant bacteria—“A Battle of the Titans”. Front Microbiol 9: 1441. Link: https://bit.ly/3k5RHBO
  61. Drulis-Kawa Z, Dorotkiewicz-Jach A (2010) Liposomes as delivery systems for antibiotics. Int J Pharm 387: 187-198. Link: https://bit.ly/2XJ1qXr
  62. Alhajlan M, Alhariri M, Omri A (2013) Efficacy and safety of liposomal clarithromycin and its effect on Pseudomonas aeruginosa virulence factors. Antimicrob Agents Chemother 57: 2694-2704. Link: https://bit.ly/3ALezxg
  63. Nkanga CI, Bapolisi AM, Okafor NI, Krause RWM (2019) General perception of liposomes: formation, manufacturing and applications. Liposomes-advances and perspectives. Link: https://bit.ly/3srIdVm
  64. Bazzaz BSF, Khameneh B, Zarei H, Golmohammadzadeh S (2016) Antibacterial efficacy of rifampin loaded solid lipid nanoparticles against Staphylococcus epidermidis biofilm. Microb Pathog 93: 137-144. Link: https://bit.ly/3iVg1ae
  65. Eleraky NE, Allam A, Hassan SB, Omar MM (2020) Nanomedicine fight against antibacterial resistance: an overview of the recent pharmaceutical innovations. Pharmaceutics 12: 142. Link: https://bit.ly/3CZWQ7a
  66. Hashizume H, Takahashi Y, Masuda T, Ohba SI, Ohishi T, et al. (2018) In vivo efficacy of β-lactam/tripropeptin C in a mouse septicemia model and the mechanism of reverse β-lactam resistance in methicillin-resistant Staphylococcus aureus mediated by tripropeptin C. The Journal of antibiotics 71: 79-85. Link: https://go.nature.com/3j0NkZG
  67. Jang S (2016) Multidrug efflux pumps in Staphylococcus aureus and their clinical implications. J Microbiol 54: 1-8. Link: https://bit.ly/3CZ1p1o
  68. Kadariya J, Smith TC, Thapaliya D (2014) Staphylococcus aureus and staphylococcal food-borne disease: an ongoing challenge in public health. Biomed Res Int 2014: 827965. Link: https://bit.ly/37T24Dm
  69. Kittler S, Greulich C, Diendorf J, Koller M, Epple M (2010) Toxicity of silver nanoparticles increases during storage because of slow dissolution under release of silver ions. Chem Mater 22: 4548-4554. Link: https://bit.ly/3xUhRMM
  70. Kumar R, Mondal K, Panda PK, Kaushik A, Abolhassani R, et al. (2020) Core–shell nanostructures: perspectives towards drug delivery applications. Journal of Materials Chemistry B 8: 8992-9027. Link: https://rsc.li/3xUsgrX
  71. Lacoma A, Usón L, Mendoza G, Sebastián V, Garcia-Garcia E, et al. (2020) Novel intracellular antibiotic delivery system against Staphylococcus aureus: cloxacillin-loaded poly (d, l-lactide-co-glycolide) acid nanoparticles. Nanomedicine15: 1189-1203.
  72. Luzum M, Sebolt J, Chopra V (2020) Catheter-Associated Urinary Tract Infection, Clostridioides difficile Colitis, Central Line–Associated Bloodstream Infection, and Methicillin-Resistant Staphylococcus aureus. Med Clin North Am 104: 663-679. Link: https://bit.ly/3iWPrNZ
© 2021 Giri VP, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
 

Help ?