Next Article in Journal
Influence of Microfibrillated Cellulose Additive on Strength, Elastic Modulus, Heat Release, and Shrinkage of Mortar and Concrete
Next Article in Special Issue
Preparation of Cationic Amphiphilic Nanoparticles with Modified Chitosan Derivatives for Doxorubicin Delivery
Previous Article in Journal
Friction Stir Welding of Thick Plates of 4Y3Gd Mg Alloy: An Investigation of Microstructure and Mechanical Properties
Previous Article in Special Issue
Mussel-Inspired Carboxymethyl Chitosan Hydrogel Coating of Titanium Alloy with Antibacterial and Bioactive Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in the Surface Functionalization of Nanomaterials for Antimicrobial Applications

College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
*
Author to whom correspondence should be addressed.
Materials 2021, 14(22), 6932; https://doi.org/10.3390/ma14226932
Submission received: 30 September 2021 / Revised: 5 November 2021 / Accepted: 10 November 2021 / Published: 16 November 2021
(This article belongs to the Special Issue Functional Materials/Surfaces in Biomedical Applications)

Abstract

:
Innovations in nanotechnology have had an immense impact on medicine, such as in drug delivery, tissue engineering, and medical devices that combat different pathogens. The pathogens that may cause biofilm-associated nosocomial diseases are multidrug-resistant (MDR) bacteria, such as Escherichia coli (E. coli), Pseudomonas aeruginosa (P. aeruginosa), Staphylococcus aureus (S. aureus), including both Gram-positive and Gram-negative bacterial species. About 65–80% of infections are caused by biofilm-associated pathogens creating a move in the international community toward developing antimicrobial therapies to eliminate such pathogenic infections. Several nanomaterials (NMs) have been discovered and significantly employed in various antipathogenic therapies. These NMs have unique properties of singlet oxygen production, high absorption of near-infrared irradiation, and reasonable conversion of light to heat. In this review, functionalized NPs that combat different pathogenic infections are introduced. This review highlights NMs that combat infections caused by multidrug-resistant (MDR) and other pathogenic microorganisms. It also highlights the biomedical application of NPs with regard to antipathogenic activities.

1. Introduction

The use of medical devices in the field of medicine for therapeutic and diagnostic purposes has high importance. However, the risks associated with their usage are not negligible, particularly when these devices are intended to be inserted inside the body for long periods of time. The most important associated problems are colonization of bacteria on medical devices after surgeries, having a significant impact on patient health and the costs associated with the treatment of infectious disease. These barriers are commonly connected with bacteria that cause contamination of surgical wounds during surgery or during the time of hospitalization, which may result in a nosocomial infection [1]. So, these infections are extremely difficult to handle as bacteria (e.g., methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE)) are more resistant to classical antibiotic therapies; particularly, medical devices, when colonized by bacteria, create a biofilm that is more dangerous for patient health [2]. Once bacteria make a coat on a hard surface, they can protect themselves by producing an extracellular matrix rich in exopolysaccharides. The bacteria colonization of the surface of medical devices or hard surfaces confer to biofilm a surface that provides a polysaccharide shelter that is resistant to our immune system [3].
The slow growth rate of bacteria and the biofilm microenvironment act together to facilitate the development of antibiotic resistance [4]. Furthermore, these infections are mainly caused by multidrug-resistant (MDR) bacteria, P. aeruginosa, E. coli, S. aureus, MRSA, VRE, Acinetobacter baumanii, and coagulase-negative Staphylococcus, which infect more than two million people annually in the United States, resulting in a significant loss of life and limb, with treatment requiring prolonged and costly therapeutic regimens [5]. The national institute of health (NIH) revealed that 65–80% of the infections are due to biofilm-associated pathogens [6]. The high risk regarding biofilm-associated nosocomial infections requires an international scientific movement to develop antimicrobial therapies to eliminate these infections [7]. For this purpose, nanotechnology is an emerging field in which different nanomaterials (NMs) act by surface plasmon resonance. Surface plasmon resonance is a powerful technique that retrieves information on the optical properties of NMs by striking incident light on its structure, which fluctuates the electrons at a specific resonance frequency and acts as a sensor to the transduced absorption band in the UV spectrum [8,9]. Furthermore, NPs can absorb near-infrared radiation (NIR), convert light to heat, and generate radicals, which highlight their therapeutic application against different pathogens [10]. These NPs are generally classified based on morphology, dimensionality, and chemical nature, and are categorized as isometric and inhomogeneous or dispersed and agglomerate [11].
This review highlights surface functionalization regarding the interaction of NPs with microbial cells, particularly how the unique characteristics of plasmon-based NPs influence their uptake pathway, NPs intracellular location and translocation, cytotoxicity, and biological application at the cellular level. The current knowledge of the physicochemical properties of NPs in light of antimicrobial activities will alleviate the effects of nosocomial diseases and combat these pathogens with their synergistic effects. These NPs combat microbial infection by mechanisms of reactive oxygen species (ROS)-based photodynamic therapy (PDT) and hypothermia-based photothermal therapy (PTT). PDT involves combating pathogenic bacteria via ROS, including superoxide anion (O2), hydrogen peroxide (H2O2), singlet oxygen (1O2), and hydroxyl radical (⋅OH) [12]. It is also widely accepted that ROS can bind and damage the cell membrane and cell wall, thus destroying the defense system of pathogenic microorganisms [13,14]. The PTT mechanism of NPs involves the conversion of light into heat, which can kill pathogens by absorbing light (NIR) and converting it into heat [15,16]. Most importantly, we must construct safe and engineered NMs against different microbial infections and explore their biomedical applications.

2. Surface Functionalization and Characteristics of NPs for Antimicrobial Activities

To impart antimicrobial activity to NMs, there are three main different strategies. The first is to functionalize the NMs or polymers using antimicrobial agents such as quaternary ammonium compounds (QACs) [17,18] through a mechanism of contact killing. This strategy has been used extensively for the improvement in the microbicidal effectiveness of various surfaces. In particular, QACs are mostly used as disinfectants and microbicidal agents, having long alkyl chains to induce strong toxicity against amoeba and fungi, as well as to envelop viruses [19,20]. The second is to fabricate nanocomposites by adding filler such as metal-based NMs, e.g., Ag NPs. Four steps are necessary to create the antimicrobial activity of Ag NPs: (1) approach the surface of bacteria; (2) disrupt the cell wall and membrane; (3) produce free radicals and ROS, which exerts toxicity and oxidative stress effects; and (4) modulate signal transduction pathways. The third is the encapsulation of antimicrobial drugs or biomolecules, e.g., gentamicin. These functionalized NMs have strong toxicity against different infections caused by MDR bacteria, fungi, and viruses [21,22].
Different techniques are used for surface functionalization of carbon materials, i.e., oxidative process and non-destructive surface modification. In oxidative processes, the oxidized NMs further act as a precursor for chemical modifications such as silanization, polymer grafting, esterification, alkylation, arylation, etc., while non-destructive surface modifications are achieved by polymers wrapping, surfactant adsorption, and encapsulating processes [23,24]. Furthermore, the main aims of NPs surface functionalization are to tackle toxicity, the clearance of immune responses, and attachment of more complex and specific ligands to enhance efficacy and specificity for purposes such as antipathogenic activities through NPs [25].
Recently, the structural and behavioral changes of microorganisms have rapidly increased. The appearance of resistant bacteria by enhancing antibiotic adaptation is a significant concern for public health; thus, many efforts are needed to promote the efficiency and efficacy of antimicrobial substances. Surface functionalization of antimicrobial NPs can improve their efficiency, and various studies have shown their potential for use against resistant bacteria [26]; it also reduces the tendency to agglomerate carbon materials and increase the interfacial adhesion of carbon–polymer through covalent or ionic bonds [27].
For the use of NMs in biomedical applications, two main characteristics must be considered: toxicity and cellular uptake [28]. In nanotechnology, biocompatibility is one of the important characteristics of NPs for biomedical purposes, which describes the properties of materials being compatible with living tissues. The most important goal is to internalize NPs to targeted cells/tissues compared to non-targeted cells and to minimize the toxicity of the NPs into cells [29,30]. NPs also have unique physical and chemical properties due to their high surface area and nanoscale size [31].

3. Antimicrobial Activities of Nanomaterials

Antimicrobial resistance (AMR) occurs when bacteria, viruses, fungi, and parasites change over time and no longer respond to medicines, making infections harder to treat and increasing the risk of disease spread, severe illness, and death. It was implicated that more than 70% of micro-organisms causing infections are resistant to one or more antimicrobial agents used as treatment to eradicate infections [32]. Considering the current situation worldwide, there is concern about controlling MDR pathogens for infectious diseases preventions. Additionally, the significant prevalence of cross-contaminations and drug overuses, which may lead to biofilm development, may cause nosocomial disease that shows more resistance to standard therapies [33]. There are several mechanisms through which antibiotic resistance is achieved in bacteria, i.e., drug penetration prevention into a cell, antibiotics target changes, enzymatic inactivation of antibiotics, and active excretion of an antibiotic from a cell [34]. Some pathogenic fungal strains are also involved in biofilm formation and are more dangerous than bacterial biofilm. These fungal strains are likely yeast and filamentous fungi, and the most studied model is Candida albicans (C. albicans). There are also other species (spp.), such as Histoplasma capsulatum, Paracoccidioides, Trichophyton, C. glabrata, C. tropicalis, C. parapsilosis, Cryptococcus spp., Malassezia spp., Trichosporon spp., Fusarium spp., Scedosporium spp., Lomentospora prolificans, and Coccidioides spp. Table 1 reveals the NMs that have activities against different MDR pathogens, antibacterials, antifungals, and antivirals that combat the biofilm-associated nosocomial pathogens mentioned above [35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73].
In this regard, developing non-conventional antimicrobial agents to prevent the aforementioned causes is under study. The rapid development of nanoscience and nanotechnology has shown promising potential for developing novel biocidal agents that would integrate with a biomaterial to prevent bacterial colonization and biofilm formation [33,90]. Metals with inherent antimicrobial properties such as silver, copper, and zinc, on the nanoscale, constitute a special class of antimicrobials that have a broad-spectrum antimicrobial nature and pose minimum toxicity to humans. Furthermore, a wide range of metals has antimicrobial activity, e.g., Ag, Al, As, Cd, Co, Cr, Cu, Fe, Ga, Hg, Mo, Mn, Ni, Pb, Sb, Te, and Zn [33,35,74,76,77,78,79,80,81,82,83,84,85,86]. These NMs have vast antipathogenic activities and are used to eradicate such pathogens that have strong resistance against normal therapies. These NPs tackle pathogens by having biocompatibility, biodegradability, reactive oxygen species (ROS) production, high absorption of NIR irradiation, and photodynamic and photothermal-conversion mechanisms, which show high performance in antipathogenic therapies.
Understanding the main role of nanotechnology and its biomedical applications, in addition to antipathogenic therapies, is also promising; the most remarkable roles of NMs are in stem-cell-based therapies, neurodegenerative diseases, anticancer treatment, and gene delivery, which are currently being researched [91]. However, there are also a few drawbacks to be considered during the use of NPs for in vivo models. Most NPs disturb cell viability, alter mitochondrial functions, increase oxidative stresses, and alter tight junction protein expression of the blood barriers [92]. Hence, novel biomaterials that inhibit microbial growth and have low toxicity would be of great significance [33].

4. Outlook and Further Perspectives

Many scientists have reported several studies on NPs and their biomedical applications according to current demands. The biomedical application of NPs remains the most attractive aspect for scientists to improve clinical outcomes. The targeted drug and gene delivery diagnostic algorithmic treatment approaches improve efficacy while reducing side effects. However, the promising role of nanotechnology in combating different pathogens is more reliable and technical compared to standard antibiotics therapies. Metal NPs, owing to their unique adaptable physical and chemical properties, have been investigated widely in biomedical research.
Metal NPs have a unique interaction with light, which provides competent means for tracking nano-complex therapeutics carriers within the body, allowing more efficient therapies with low adverse effects compared to current conventional therapies. Interestingly, due to their high drug payload, surface chemistry, electrostatic charges, and photothermal behavior, metal NPs enhance therapeutics’ antimicrobial drug efficacy.
In this review, we discuss the promising role of functionalizing NPs with an antimicrobial agent, adding filler on its surface or encapsulating the antimicrobial agent, which allows for combating activities against different pathogens such as MDR, bacterial, fungal, and viral infections. Conclusively, based on the current literature on pre-clinical trials, plasmon-based metal NPs and their combination approaches are good prospects in microbial infectious therapies; for instance, biocompatibility, generation of singlet oxygen, and photothermal conversion properties of plasmon-based metal NPs are useful, and helpful in anticancer therapies. Further extensive research on NPs into their long-term toxicity is essential for successful clinical use in biomedical applications. We also think that the development of simple and low-cost inorganic antimicrobial agents such as NPs as alternatives for traditional antimicrobial agents might be promising for the future of the pharmaceutical, food, and medical industries.

Author Contributions

S.S.K.; Conceptualization, writing—original draft preparation, I.U.; writing—review and editing, S.U.; validation, R.A.; formal analysis, H.X.; Conceptualization, K.N.; investigation, C.L.; project administration, L.L.; supervision, funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by National Natural Science Foundation of China (grant number 52073022).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are avalible on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schierholz, J.M.; Beuth, J. Implant infections: A haven for opportunistic bacteria. J. Hosp. Infect. 2001, 49, 87–93. [Google Scholar] [CrossRef]
  2. Dupont, H. The empiric treatment of nosocomial intra-abdominal infections. Int. J. Infect. Dis. 2007, 11, S1–S6. [Google Scholar] [CrossRef] [Green Version]
  3. Pérez-Köhler, B.; Bayon, Y.; Bellón, J.M. Mesh infection and hernia repair: A review. Surg. Infect. 2016, 17, 124–137. [Google Scholar] [CrossRef]
  4. Olsen, I. Biofilm-specific antibiotic tolerance and resistance. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 877–886. [Google Scholar] [CrossRef]
  5. De Miguel, I.; Prieto, I.; Albornoz, A.; Sanz, V.; Weis, C.; Turon, P.; Quidant, R. Plasmon-based biofilm inhibition on surgical implants. Nano Lett. 2019, 19, 2524–2529. [Google Scholar] [CrossRef] [PubMed]
  6. Jamal, M.; Ahmad, W.; Andleeb, S.; Jalil, F.; Imran, M.; Nawaz, M.A.; Hussain, T.; Ali, M.; Rafiq, M.; Kamil, M.A. Bacterial biofilm and associated infections. J. Chin. Med. Assoc. 2018, 81, 7–11. [Google Scholar] [CrossRef] [PubMed]
  7. Del Pozo, J.L.; Patel, R. The challenge of treating biofilm-associated bacterial infections. Clin. Pharmacol. Ther. 2007, 82, 204–209. [Google Scholar] [CrossRef] [PubMed]
  8. Ghosh, S.K.; Pal, T. Interparticle coupling effect on the surface plasmon resonance of gold nanoparticles: From theory to applications. Chem. Rev. 2007, 107, 4797–4862. [Google Scholar] [CrossRef]
  9. Olson, J.; Dominguez-Medina, S.; Hoggard, A.; Wang, L.Y.; Chang, W.S.; Link, S. Optical characterization of single plasmonic nanoparticles. Chem. Soc. Rev. 2015, 44, 40–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Jaque, D.; Maestro, L.M.; del Rosal, B.; Haro-Gonzalez, P.; Benayas, A.; Plaza, J.L.; Rodríguez, E.M.; Solé, J.G. Nanoparticles for photothermal therapies. Nanoscale 2014, 6, 9494–9530. [Google Scholar] [CrossRef]
  11. Saleh, T.A. Nanomaterials: Classification, properties, and environmental toxicities. Environ. Technol. Innov. 2020, 1–16. [Google Scholar] [CrossRef]
  12. Ren, Y.; Liu, H.; Liu, X.; Zheng, Y.; Li, Z.; Li, C.; Yeung, K.W.K.; Zhu, S.; Liang, Y.; Cui, Z.; et al. Photoresponsive materials for antibacterial applications. Cell Rep. Phys. Sci. 2020, 1, 1–30. [Google Scholar] [CrossRef]
  13. Wei, T.; Yu, Q.; Chen, H. Responsive and synergistic antibacterial coatings: Fighting against bacteria in a smart and effective way. Adv. Healthc. Mater. 2019, 8. [Google Scholar] [CrossRef]
  14. Li, J.; Liu, X.; Tan, L.; Liang, Y.; Cui, Z.; Yang, X.; Zhu, S.; Li, Z.; Zheng, Y.; Yeung, K.W.K.; et al. Light-activated rapid disinfection by accelerated charge transfer in red phosphorus/ZnO heterointerface. Smal. Meth. 2019, 3. [Google Scholar] [CrossRef]
  15. Chen, B.Q.; Kankala, R.K.; Zhang, Y.; Xiang, S.T.; Tang, H.X.; Wang, Q.; Yang, D.Y.; Wang, S.B.; Zhang, Y.S.; Liu, G.; et al. Gambogic acid augments black phosphorus quantum dots (BPQDs)-based synergistic chemo-photothermal therapy through downregulating heat shock protein expression. Chem. Eng. Sci. 2020, 390, 5302–5318. [Google Scholar] [CrossRef]
  16. Xu, J.W.; Yao, K.; Xu, Z.K. Nanomaterials with a photothermal effect for antibacterial activities: An overview. Nanoscale 2019, 11, 8680–8691. [Google Scholar] [CrossRef]
  17. Chaker, A.; Boufi, S. Cationic nanofibrillar cellulose with high antibacterial properties. Carbohydr. Polym. 2015, 131, 224–232. [Google Scholar] [CrossRef]
  18. Makvandi, P.; Jamaledin, R.; Jabbari, M.; Nikfarjam, N.; Borzacchiello, A. Antibacterial quaternary ammonium compounds in dental materials: A systematic review. Dent. Mater. 2018, 34, 851–867. [Google Scholar] [CrossRef]
  19. Makvandi, P.; Gu, J.T.; Zare, E.N.; Ashtari, B.; Moeini, A.; Tay, F.R.; Niu, L.N. Polymeric and inorganic nanoscopical antimicrobial fillers in dentistry. Acta Biomater. 2020, 101, 69–101. [Google Scholar] [CrossRef]
  20. Makvandi, P.; Pollini, M.; Gallo, A.L.; Maffezzoli, A.; Esposito Corcione, C.; Montagna, F.; Paladini, F.; Jamaledin, R. Antimicrobial modified hydroxyapatite composite dental bite by stereolithography. Polym. Adv. Technol. 2017, 29, 364–371. [Google Scholar] [CrossRef]
  21. Dakal, T.C.; Kumar, A.; Majumdar, R.S.; Yadav, V. Mechanistic basis of antimicrobial actions of silver nanoparticles. Front. Microbiol. 2016, 7, 1831. [Google Scholar] [CrossRef] [Green Version]
  22. Delfi, M.; Ghomi, M.; Zarrabi, A.; Mohammadinejad, R.; Taraghdari, Z.B.; Ashrafizadeh, M.; Zare, E.N.; Agarwal, T.; Padil, V.V.; Mokhtari, B.; et al. Functionalization of polymers and nanomaterials for biomedical applications: Antimicrobial platforms and drug carriers. Prosthe 2020, 2, 117–139. [Google Scholar] [CrossRef]
  23. Sham, M.L.; Kim, J.K. Surface functionalities of multi-wall carbon nanotubes after UV/Ozone and TETA treatments. Carbon 2006, 44, 768–777. [Google Scholar] [CrossRef]
  24. Kostarelos, K.; Lacerda, L.; Pastorin, G.; Wu, W.; Wieckowski, S.; Luangsivilay, J.; Godefroy, S.; Pantarotto, D.; Briand, J.P.; Muller, S.; et al. Cellular uptake of functionalized carbon nanotubes is independent of functional group and cell type. Nat. Nanotechnol. 2007, 2, 108–113. [Google Scholar] [CrossRef]
  25. Sanità, G.; Carrese, B.; Lamberti, A. Nanoparticle surface functionalization: How to improve biocompatibility and cellular internalization. Front. Mol. Biosci. 2020, 7, 381. [Google Scholar] [CrossRef]
  26. Bahrami, A.; Delshadi, R.; Jafari, S.M. Active delivery of antimicrobial nanoparticles into microbial cells through surface functionalization strategies. Trends Food Sci. Technol. 2020, 99, 217–228. [Google Scholar] [CrossRef]
  27. Nayak, L.; Rahaman, M.; Giri, R. Surface modification/functionalization of carbon materials by different techniques: An overview. Carbon-Contain. Polym. Compos. 2019, 65–98. [Google Scholar] [CrossRef]
  28. Santos-Rasera, J.R.; Neto, A.S.A.; Monteiro, R.T.R.; van Gestel, C.A.; de Carvalho, H.W.P. Toxicity, bioaccumulation and biotransformation of Cu oxide nanoparticles in Daphnia magna. Environ. Sci. Nano 2019, 6, 2897–2906. [Google Scholar] [CrossRef]
  29. Liu, T.; Bai, R.; Zhou, H.; Wang, R.; Liu, J.; Zhao, Y.; Chen, C. The effect of size and surface ligands of iron oxide nanoparticles on blood compatibility. RSC Adv. 2020, 10, 7559–7569. [Google Scholar] [CrossRef]
  30. Bharadwaj, A. An Overview on Biomaterials and Its Applications in Medical Science. Mater. Sci. Eng. 2021, 1116, 012178. [Google Scholar] [CrossRef]
  31. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  32. Malarkodi, C.; Rajeshkumar, S.; Paulkumar, K.; Vanaja, M.; Gnanajobitha, G.; Annadurai, G. Biosynthesis and antimicrobial activity of semiconductor nanoparticles against oral pathogens. Bioinorg. Chem. Appl. 2014, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Agnihotri, S.; Dhiman, N.K. Development of nano-antimicrobial bio=materials for biomedical applications. Adv. Bio. Mat. For. Biomed. Appl. 2017, 66, 479–545. [Google Scholar] [CrossRef]
  34. Ahamed, M.N.; Sankar, S.; Kashif, P.M.; Basha, S.H.; Sastry, T.P. Evaluation of biomaterial containing regenerated cellulose and chitosan incorporated with silver nanoparticles. Int. J. Biol. 2015, 72, 680–686. [Google Scholar] [CrossRef] [PubMed]
  35. Costa-Orlandi, C.B.; Sardi, J.C.; Pitangui, N.S.; de Oliveira, H.C.; Scorzoni, L.; Galeane, M.C.; Medina-Alarcón, K.P.; Melo, W.C.; Marcelino, M.Y.; Braz, J.D.; et al. Fungal biofilms and polymicrobial diseases. J. Fungi 2017, 3, 22. [Google Scholar] [CrossRef]
  36. Feng, Q.L.; Wu, J.; Chen, G.Q.; Cui, F.Z.; Kim, T.N.; Kim, J.O. A mechanistic study of the antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus. J. Biomed. Mater. Res. 2000, 52, 662–668. [Google Scholar] [CrossRef]
  37. Jung, W.K.; Koo, H.C.; Kim, K.W.; Shin, S.; Kim, S.H.; Park, Y.H. Antibacterial activity and mechanism of action of the silver ion in Staphylococcus aureus and Escherichia coli. Appl. Environ. Microbiol. 2008, 74, 2171–2178. [Google Scholar] [CrossRef] [Green Version]
  38. Nanda, A.; Saravanan, M. Biosynthesis of silver nanoparticles from Staphylococcus aureus and its antimicrobial activity against MRSA and MRSE. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 452–456. [Google Scholar] [CrossRef]
  39. Kim, J.S.; Kuk, E.; Yu, K.N.; Kim, J.H.; Park, S.J.; Lee, H.J.; Kim, S.H.; Park, Y.K.; Park, Y.H.; Hwang, C.Y.; et al. Antimicrobial effects of silver nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2007, 3, 95–101. [Google Scholar] [CrossRef]
  40. Gil-Tomás, J.; Tubby, S.; Parkin, I.P.; Narband, N.; Dekker, L.; Nair, S.P.; Wilson, M.; Street, C. Lethal photosensitisation of Staphylococcus aureus using a toluidine blue O–tiopronin–gold nanoparticle conjugate. J. Mater. Chem. 2007, 17, 3739–3746. [Google Scholar] [CrossRef]
  41. MubarakAli, D.; Thajuddin, N.; Jeganathan, K.; Gunasekaran, M. Plant extract mediated synthesis of silver and gold nanoparticles and its antibacterial activity against clinically isolated pathogens. Colloids Surf. B Biointerfaces 2011, 85, 360–365. [Google Scholar] [CrossRef] [PubMed]
  42. Matsunaga, T.; Tomoda, R.; Nakajima, T.; Nakamura, N.O.; Komine, T. Continuous-sterilization system that uses photo semiconductor powders. Appl. Environ. Microbiol. 1988, 54, 1330–1333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Othman, S.H.; Abd, S.N.R.; Zainal, N.; Kadir, B.R.; Talib, R.A. Antimicrobial activity of TiO2 nanoparticle-coated film for potential food packaging applications. Int. J. Photoenergy 2014, 1–6. [Google Scholar] [CrossRef] [Green Version]
  44. Ren, G.; Hu, D.; Cheng, E.W.; Vargas-Reus, M.A.; Reip, P.; Allaker, R.P. Characterization of copper oxide nanoparticles for antimicrobial applications. Int. J. Antimicrob. Agents 2009, 33, 587–590. [Google Scholar] [CrossRef] [PubMed]
  45. Richards, R.; Li, W.; Decker, S.; Davidson, C.; Koper, O.; Zaikovski, V.; Volodin, A.; Rieker, T.; Klabunde, K.J. Consolidation of metal oxide nanocrystals. Reactive pellets with controllable pore structure that represent a new family of porous, inorganic materials. J. Am. Chem. Soc. 2000, 122, 4921–4925. [Google Scholar] [CrossRef]
  46. Krishnamoorthy, K.; Manivannan, G.; Kim, S.J.; Jeyasubramanian, K.; Premanathan, M. Antibacterial activity of MgO nanoparticles based on lipid peroxidation by oxygen vacancy. J. Nanoparticle Res. 2012, 14, 1–10. [Google Scholar] [CrossRef]
  47. Roy, A.; Gauri, S.S.; Bhattacharya, M.; Bhattacharya, J. Antimicrobial activity of CaO nanoparticles. J. Biomed. Nanotech. 2013, 9, 1570–1578. [Google Scholar] [CrossRef] [PubMed]
  48. Prashanth, P.A.; Raveendra, R.S.; Hari, K.R.; Ananda, S.; Bhagya, N.P.; Nagabhushana, B.M.; Lingaraju, K.; Raja, N.H. Synthesis, characterizations, antibacterial and photoluminescence studies of solution combustion-derived α-Al2O3 nanoparticles. J. Asian Ceram. Soc. 2015, 3, 345–351. [Google Scholar] [CrossRef] [Green Version]
  49. Jastrzębska, A.M.; Radziun, E.; Rosłon, M.; Kunicki, A.R.; Olszyna, A.R.; Dudkiewicz-Wilczyńska, J.; Anuszewska, E.; Karwowska, E. In vitro assessment of antibacterial properties and cytotoxicity of Al2O3–Ag nanopowders. Adv. Appl. Ceram. 2011, 110, 353–359. [Google Scholar] [CrossRef]
  50. Khezerlou, A.; Alizadeh-Sani, M.; Azizi-Lalabadi, M.; Ehsani, A. Nanoparticles and their antimicrobial properties against pathogens including bacteria, fungi, parasites and viruses. Microb. Pathog. 2018, 123, 505–526. [Google Scholar] [CrossRef] [PubMed]
  51. Silvestry-Rodriguez, N.; Sicairos-Ruelas, E.E.; Gerba, C.P.; Bright, K.R. Silver as a disinfectant. Rev. Environ. Contam. Toxicol. 2007, 23–45. [Google Scholar] [CrossRef]
  52. Bagchi, B.; Kar, S.; Dey, S.K.; Bhandary, S.; Roy, D.; Mukhopadhyay, T.K.; Das, S.; Nandy, P. In situ synthesis and antibacterial activity of copper nanoparticle loaded natural montmorillonite clay based on contact inhibition and ion release. Colloids Surf. B Biointerfaces 2013, 108, 358–365. [Google Scholar] [CrossRef] [PubMed]
  53. Panáček, A.; Kolář, M.; Večeřová, R.; Prucek, R.; Soukupova, J.; Kryštof, V.; Hamal, P.; Zbořil, R.; Kvitek, L. Antifungal activity of silver nanoparticles against Candida spp. Biomaterials 2009, 30, 6333–6340. [Google Scholar] [CrossRef] [PubMed]
  54. Jalal, M.; Ansari, M.A.; Alzohairy, M.A.; Ali, S.G.; Khan, H.M.; Almatroudi, A.; Raees, K. Biosynthesis of silver nanoparticles from oropharyngeal Candida glabrata isolates and their antimicrobial activity against clinical strains of bacteria and fungi. Nanomaterials 2018, 8, 586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Lipovsky, A.; Nitzan, Y.; Gedanken, A.; Lubart, R. Antifungal activity of ZnO nanoparticles—The role of ROS mediated cell injury. Nanotechnology 2011, 22, 1–5. [Google Scholar] [CrossRef]
  56. Zaka, M.; Hashmi, S.S.; Siddiqui, M.A.; Rahman, L.; Mushtaq, S.; Ali, H.; Hano, C.; Abbasi, B.H. Callus-mediated biosynthesis of Ag and ZnO nanoparticles using aqueous callus extract of Cannabis sativa: Their cytotoxic potential and clinical potential against human pathogenic bacteria and fungi. Green Process. Synth. 2021, 10, 569–584. [Google Scholar] [CrossRef]
  57. Gómez-Ortíz, N.; de la Rosa-García, S.; González-Gómez, W.; Soria-Castro, M.; Quintana, P.; Oskam, G.; Ortega-Morales, B. Antifungal coatings based on Ca(OH)2 mixed with ZnO/TiO2 nanomaterials for protection of limestone monuments. Appl. Mater. Interfaces 2013, 5, 1556–1565. [Google Scholar] [CrossRef] [PubMed]
  58. Rajeswari, V.D.; Eed, E.M.; Elfasakhany, A.; Badruddin, I.A.; Kamangar, S.; Brindhadevi, K. Green synthesis of titanium dioxide nanoparticles using Laurus nobilis (bay leaf): Antioxidant and antimicrobial activities. Appl. Nanosci. 2021, 1–8. [Google Scholar] [CrossRef]
  59. Giannousi, K.; Sarafidis, G.; Mourdikoudis, S.; Pantazaki, A.; Dendrinou-Samara, C. Selective synthesis of Cu2O and Cu/Cu2O NPs: Antifungal activity to yeast saccharomyces cerevisiae and DNA interaction. Inorg. Chemist. 2014, 53, 9657–9666. [Google Scholar] [CrossRef]
  60. Pugazhendhi, A.; Kumar, S.S.; Manikandan, M.; Saravanan, M. Photocatalytic properties and antimicrobial efficacy of Fe doped CuO nanoparticles against the pathogenic bacteria and fungi. Microb. Pathog. 2018, 122, 84–89. [Google Scholar] [CrossRef]
  61. Sawai, J.; Yoshikawa, T. Measurement of fungi by an indirect conductimetric assay. Lett. Appl. Microbiol. 2003, 37, 40–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Abdel-Aziz, M.M.; Emam, T.M.; Elsherbiny, E.A. Bioactivity of magnesium oxide nanoparticles synthesized from cell filtrate of endobacterium Burkholderia rinojensis against Fusarium oxysporum. Mater. Sci. Eng. 2020, 109, 1–10. [Google Scholar] [CrossRef]
  63. Maringgal, B.; Hashim, N.; Tawakkal, I.S.M.A.; Hamzah, M.H.; Mohamed, M.T.M. Biosynthesis of CaO nanoparticles using Trigona sp. Honey: Physicochemical characterization, antifungal activity, and cytotoxicity properties. J. Mater. Res. Technol. 2020, 9, 11756–11768. [Google Scholar] [CrossRef]
  64. Jayaseelan, C.; Ramkumar, R.; Rahuman, A.A.; Perumal, P. Green synthesis of gold nanoparticles using seed aqueous extract of Abelmoschus esculentus and its antifungal activity. Ind. Crops Prod. 2013, 45, 423–429. [Google Scholar] [CrossRef]
  65. Bhuyan, B.; Paul, A.; Paul, B.; Dhar, S.S.; Dutta, P. Paederia foetida Linn. promoted biogenic gold and silver nanoparticles: Synthesis, characterization, photocatalytic and in vitro efficacy against clinically isolated pathogens. J. Photochem. Photobiol. 2017, 173, 210–215. [Google Scholar] [CrossRef] [PubMed]
  66. Jalal, M.; Ansari, M.A.; Shukla, A.K.; Ali, S.G.; Khan, H.M.; Pal, R.; Alam, J.; Cameotra, S.S. Green synthesis and antifungal activity of Al2O3 NPs against fluconazole-resistant Candida spp isolated from a tertiary care hospital. RSC Adv. 2016, 6, 107577–107590. [Google Scholar] [CrossRef]
  67. Piroonpan, T.; Huajaikaew, E.; Katemake, P.; Pasanphan, W. Surface modification of SiO2 nanoparticles with PDMAEMA brushes and Ag nanoparticles as antifungal coatings using electron beam assisted synthesis. Mater. Chem. Phys. 2020, 253, 123438. [Google Scholar] [CrossRef]
  68. Ismail, A.; Kabary, H.; Samy, A. Synthesis of α-Al2O3 Nanoparticles from Pepsi Cans Wastes and Its Fungicidal Effect on Some Mycotoxins Producing Fungal Isolates. Res. Squar. 2021. [Google Scholar] [CrossRef]
  69. Lara, H.H.; Ayala-Nuñez, N.V.; Ixtepan-Turrent, L.; Rodriguez-Padilla, C. Mode of antiviral action of silver nanoparticles against HIV-1. J. Nanobiotech. 2010, 8, 1–10. [Google Scholar] [CrossRef]
  70. Baram-Pinto, D.; Shukla, S.; Perkas, N.; Gedanken, A.; Sarid, R. Inhibition of herpes simplex virus type 1 infection by silver nanoparticles capped with mercaptoethane sulfonate. Bioconjug. Chem. 2009, 20, 1497–1502. [Google Scholar] [CrossRef]
  71. El-Sheekh, M.M.; Shabaan, M.T.; Hassan, L.; Morsi, H.H. Antiviral activity of algae biosynthesized silver and gold nanoparticles against Herps Simplex (HSV-1) virus in vitro using cell-line culture technique. Int. J. Environ. Health Res. 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  72. Tremiliosi, G.C.; Simoes, L.G.P.; Minozzi, D.T.; Santos, R.I.; Vilela, D.C.; Durigon, E.L.; Machado, R.R.G.; Medina, D.S.; Ribeiro, L.K.; Rosa, I.L.V.; et al. Ag nanoparticles-based antimicrobial polycotton fabrics to prevent the transmission and spread of SARS-CoV-2. BioRxiv 2020. [Google Scholar] [CrossRef]
  73. Antunes Filho, S.; dos Santos, O.A.L.; dos Santos, M.S.; Backx, B.P. Exploiting nanotechnology to target viruses. J. Nanotech. Nanomater. 2020, 1, 11–15. [Google Scholar]
  74. Chmielewska, S.J.; Skłodowski, K.; Depciuch, J.; Deptuła, P.; Piktel, E.; Fiedoruk, K.; Kot, P.; Paprocka, P.; Fortunka, K.; Wollny, T.; et al. Bactericidal properties of rod-, peanut-, and star-shaped gold nanoparticles coated with ceragenin CSA-131 against multidrug-resistant bacterial strains. Pharmaceutics 2021, 13, 425. [Google Scholar] [CrossRef] [PubMed]
  75. Mickymaray, S. One-step synthesis of silver nanoparticles using Saudi Arabian desert seasonal plant Sisymbrium irio and antibacterial activity against multidrug-resistant bacterial strains. Biomolecules 2019, 9, 662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Hamed, S.; Emara, M.; Shawky, R.M.; El-domany, R.A.; Youssef, T. Silver nanoparticles: Antimicrobial activity, cytotoxicity, and synergism with N-acetyl cysteine. J. Basic Microbiol. 2017, 57, 659–668. [Google Scholar] [CrossRef] [PubMed]
  77. Alharbi, F.A.; Alarfaj, A.A. Green synthesis of silver nanoparticles from Neurada procumbens and its antibacterial activity against multi-drug resistant microbial pathogens. J. King Saud. Univ. Sci. 2020, 32, 1346–1352. [Google Scholar] [CrossRef]
  78. Saravanan, M.; Barik, S.K.; MubarakAli, D.; Prakash, P.; Pugazhendhi, A. Synthesis of silver nanoparticles from Bacillus brevis (NCIM 2533) and their antibacterial activity against pathogenic bacteria. Microb. Pathog. 2018, 116, 221–226. [Google Scholar] [CrossRef]
  79. Saeed, S.; Iqbal, A.; Ashraf, M.A. Bacterial-mediated synthesis of silver nanoparticles and their significant effect against pathogens. Environ. Sci. Pollut. Res. 2020, 27, 37347–37356. [Google Scholar] [CrossRef]
  80. Ebrahim-Saraie, H.S.; Heidari, H.; Rezaei, V.; Mortazavi, S.M.J.; Motamedifar, M. Promising antibacterial effect of copper oxide nanoparticles against several multidrug resistant uropathogens. Pharm. Sci. 2018, 24, 213–218. [Google Scholar] [CrossRef]
  81. Sen, S.; Sarkar, K. Effective Biocidal and Wound Healing Cogency of Biocompatible Glutathione: Citrate-Capped Copper Oxide Nanoparticles against Multidrug-Resistant Pathogenic Enterobacteria. Microb. Drug Resist. 2021, 27, 616–627. [Google Scholar] [CrossRef]
  82. El-Sayyad, G.S.; El-Bastawisy, H.S.; Gobara, M.; El-Batal, A.I. Gentamicin-assisted mycogenic selenium nanoparticles synthesized under gamma irradiation for robust reluctance of resistant urinary tract infection-causing pathogens. Biol. Trace Elem. Res. 2020, 195, 323–342. [Google Scholar] [CrossRef] [PubMed]
  83. Al Edhari, B.; Mashreghi, M.; Makhdoumi, A.; Darroudi, M. Antibacterial and antibiofilm efficacy of Ag NPs, Ni NPs and Al2O3 NPs singly and in combination against multidrug-resistant Klebsiella pneumoniae isolates. J. Trace Elem. Med. Biol. 2021, 68, 1–13. [Google Scholar] [CrossRef]
  84. Ali, S.S.; Sonbol, F.I.; Sun, J.; Hussein, M.A.; Hafez, A.E.E.; Abdelkarim, E.A.; Kornaros, M.; Ali, A.; Azab, M. Molecular characterization of virulence and drug resistance genes-producing Escherichia coli isolated from chicken meat: Metal oxide nanoparticles as novel antibacterial agents. Microb. Pathog. 2020, 143, 104164. [Google Scholar] [CrossRef] [PubMed]
  85. Ali, S.S.; Moawad, M.S.; Hussein, M.A.; Azab, M.; Abdelkarim, E.A.; Badr, A.; Sun, J.; Khalil, M. Efficacy of metal oxide nanoparticles as novel antimicrobial agents against multi-drug and multi-virulent Staphylococcus aureus isolates from retail raw chicken meat and giblets. Int. J. Food Microbiol. 2021, 344. [Google Scholar] [CrossRef] [PubMed]
  86. Bellio, P.; Luzi, C.; Mancini, A.; Cracchiolo, S.; Passacantando, M.; di Pietro, L.; Perilli, M.; Amicosante, G.; Santucci, S.; Celenza, G. Cerium oxide nanoparticles as potential antibiotic adjuvant. Effects of CeO2 nanoparticles on bacterial outer membrane permeability. Biochim. Biophys. Acta Biomembr. 2018, 1860, 2428–2435. [Google Scholar] [CrossRef] [PubMed]
  87. Ullah, S.; Khan, S.S.; Ren, Y.; Zhang, X.; Qin, M.; Xiong, X.; Krastev, R.; Jan, A.U.; Liu, L.; Yuan, Q. Near-infrared laser 808-nm excitable palladium nano-dots loaded on graphene oxide hybrid for the antibacterial activity. Appl. Organomet. Chem. 2021, 35, 1–11. [Google Scholar] [CrossRef]
  88. Ling, Y.; Luo, Y.; Luo, J.; Wang, X.; Sun, R. Novel antibacterial paper based on quaternized carboxymethyl chitosan/organic montmorillonite/Ag NP nanocomposites. Ind. Crops Prod. 2013, 51, 470–479. [Google Scholar] [CrossRef]
  89. Chai, W.; Zakrzewski, S.S.; Günzel, D.; Pieper, R.; Wang, Z.; Twardziok, S.; Janczyk, P.; Osterrieder, N.; Burwinkel, M. High-dose dietary zinc oxide mitigates infection with transmissible gastroenteritis virus in piglets. BMC Vet. Res. 2014, 10, 1–10. [Google Scholar] [CrossRef] [Green Version]
  90. Qi, H.; Shan, P.; Wang, Y.; Li, P.; Wang, K.; Yang, L. Nanomedicines for the Efficient Treatment of Intracellular Bacteria: The “ART” Principle. Front. Chem. 2021, 9, 1–9. [Google Scholar] [CrossRef] [PubMed]
  91. Dong, Y.; Wu, X.; Chen, X.; Zhou, P.; Xu, F.; Liang, W. Nanotechnology shaping stem cell therapy: Recent advances, application, challenges, and future outlook. Biomed. Pharmacother. 2021, 137, 1–9. [Google Scholar] [CrossRef] [PubMed]
  92. Bahadar, H.; Maqbool, F.; Niaz, K.; Abdollahi, M. Toxicity of nanoparticles and an overview of current experimental models. Iran. Biomed. J. 2016, 20, 1. [Google Scholar] [PubMed]
Table 1. Activities of different NPs against MDR-pathogens, antibacterial, antifungal and antiviral.
Table 1. Activities of different NPs against MDR-pathogens, antibacterial, antifungal and antiviral.
NPsActivityTargeted PathogenRefs.
Au NPsMDR pathogensMethicillin-resistant S. Aureus, various groups of MDR (multidrug-resistant) Gram-positive (MRSA, MRSE, and MLSB), and Gram-negative (extended-spectrum betalactamase (ESBL), AmpC, and CR) pathogens[32,74]
Ag NPsS. epidermidis, MRSA, VRE, ESBL-producing organisms, MDR E. coli, P. aeruginosa, K. pneumoniae, carbapenem- and polymyxin B-resistant A. baumannii, Salmonella typhi (S. typhi), and S. aureus Carbapenem-resistant P. aeruginosa and carbapenem-resistant Enterobacteriaceae (CRE)[32,75,76,77,78,79]
Cu NPsS. aureus, E. faecalis, E. coli, and P. aeruginosa, K. quasipneumoniae and Enterobacter sp.[80,81]
Se NPsS. aureus, P. aeruginosa, and E. coli[82]
Al NPsMDR K. pneumoniae[83]
Metal oxide NPs
ZnO2-NPs
ZnO NPs
TiO2 NPs
MDR E. coli, S. aureus[84,85]
CeO2 NPsK. pneumoniae[86]
Antibacterial activities of different NPs
Ag NPsAntibacterialE. coli, B. subtilis, S. aureus, methicillin-resistant coagulase-negative Staphylococci,
VRE faecium, ESBL-positive K. Pneumonia, S. typhi, Vibrio cholera
[36,37,38,39]
Au NPsMRSA, VRE-faecium, E. coli, P. aeruginosa[40]
TiO2 NPsE. coli 0157:H7, S. aureus,
L. monocytogenes S. enteritidis,
P. fluorescens
[41]
ZnO NPsE. coli 0157:H7, B. subtilis, P. fluorescens,
L. monocytogenes, S. enteritidis, S. aureus,
S. typhimurium
[42,43]
CuO NPsB. subtilis, L. monocytogenes, S. aureus,
E. coli
[44]
HSA-GO-PdE. coli[87]
MgO NPsB. subtilis, E. coli, S. aureus, B. megaterium[45,46]
CaO NPsS. aureus, S. epidermidis, E. coli, S. mutans[47]
Al2O3 NPsE. coli, P. aeruginosa, S. aureus, B. subtilis,
K. aerogenes, P. desmolyticum
[48,49,50]
SiO2 NPsE. coli, S. mutans, B. subtilis[51]
Clay NPsE. coli, E. faecalis, S. aureus,
P. aeruginosa
[52]
Antifungal activities of different NPs
Ag NPsAntifungalC. albicans,T. mentagrophyts, B. sorokiniana, M. grisea[53,54,55]
ZnO NPsB. cinerea, P. expansum, A. flavus, S. cerevisiae, C. albicans, R. stolonifera, F. oxysporum, Mucor, A. fumigatus, A. niger, and F. solani[50,54,55,56]
TiO2 NPsCandida. spp. P. Expansum, A. niger spp.
P. oxalicum
[57,58]
CuO NPsA. niger, Rhizopus oryzae, A. flavus,
Cladosporium carrionii, Mucor,
S. cerevisiae, P. notatum, C. albicans
[59,60]
MgO NPs Saccharomyces cerevisiae, C. albicans,
A. niger, R. stolonifer, Fusarium oxysporum f. sp. lycopersici
[61,62]
CaO NPsS. cerevisiae, C. albicans, A. Niger, R. stolonifer, C. brevisporum[61,63]
Au NPsPuccinia graminis tritci, A. flavus, A. niger C. albicans[64,65]
SiO2 NPsCandida. spp., Dermatophytes spp., A. niger, and S. racemosum[66,67]
Al2O3 NPs Candida. spp., S. quadricauda, A. niger, A. flavus, Fusarium spp., and Alternaria spp.[66,68]
Antiviral activities of different NPs
Au NPsAntiviralHIV virus, Influenza virus, Herpes Simplex virus (HSV-1)[69,70,71]
Ag NPs,
Ag2O|AgO-NPs
HIV-1, Influenza virus, Herpes Simplex virus, Respiratory syncytial virus, Monkey pox virus, SARS-CoV-2[69,70,71,72,88,89]
TiO2 NPsInactivates bacteriophages[50]
CuI NPsInfluenza A virus, feline Calicivirus (FCV)[73]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Khan, S.S.; Ullah, I.; Ullah, S.; An, R.; Xu, H.; Nie, K.; Liu, C.; Liu, L. Recent Advances in the Surface Functionalization of Nanomaterials for Antimicrobial Applications. Materials 2021, 14, 6932. https://doi.org/10.3390/ma14226932

AMA Style

Khan SS, Ullah I, Ullah S, An R, Xu H, Nie K, Liu C, Liu L. Recent Advances in the Surface Functionalization of Nanomaterials for Antimicrobial Applications. Materials. 2021; 14(22):6932. https://doi.org/10.3390/ma14226932

Chicago/Turabian Style

Khan, Shahin Shah, Irfan Ullah, Sadeeq Ullah, Ruipeng An, Haijun Xu, Kaili Nie, Chaoyong Liu, and Luo Liu. 2021. "Recent Advances in the Surface Functionalization of Nanomaterials for Antimicrobial Applications" Materials 14, no. 22: 6932. https://doi.org/10.3390/ma14226932

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop